EP1765362B1 - Compositions and methods for treatment of neovascular diseases - Google Patents

Compositions and methods for treatment of neovascular diseases Download PDF

Info

Publication number
EP1765362B1
EP1765362B1 EP05804888A EP05804888A EP1765362B1 EP 1765362 B1 EP1765362 B1 EP 1765362B1 EP 05804888 A EP05804888 A EP 05804888A EP 05804888 A EP05804888 A EP 05804888A EP 1765362 B1 EP1765362 B1 EP 1765362B1
Authority
EP
European Patent Office
Prior art keywords
trprs
seq
compound
angiostatic
mice
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP05804888A
Other languages
German (de)
French (fr)
Other versions
EP1765362A2 (en
EP1765362A4 (en
Inventor
Martin Friedlander
Hilda Edith Aguilar
Michael I. Dorrell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Scripps Research Institute
Original Assignee
Scripps Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Scripps Research Institute filed Critical Scripps Research Institute
Priority to PL12002028T priority Critical patent/PL2484365T3/en
Priority to EP12002027.6A priority patent/EP2484364B1/en
Priority to DK12002028.4T priority patent/DK2484365T3/en
Priority to PL05804888T priority patent/PL1765362T3/en
Priority to EP12002028.4A priority patent/EP2484365B1/en
Publication of EP1765362A2 publication Critical patent/EP1765362A2/en
Publication of EP1765362A4 publication Critical patent/EP1765362A4/en
Application granted granted Critical
Publication of EP1765362B1 publication Critical patent/EP1765362B1/en
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/5381,4-Oxazines, e.g. morpholine ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/93Ligases (6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y601/00Ligases forming carbon-oxygen bonds (6.1)
    • C12Y601/01Ligases forming aminoacyl-tRNA and related compounds (6.1.1)
    • C12Y601/01002Tryptophan-tRNA ligase (6.1.1.2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • compositions comprising a combination of angiostatic and antiangiogenic drugs said compositions being useful in the treatment of neovascular diseases, such as retinal neovascular diseases.
  • age related macular degeneration affects 12-15 million Americans over the age of 65 and causes visual loss in 10-15% of them as a direct effects of choroidal (sub-retinal) neovascularization.
  • the leading cause of visual loss for Americans under the age of 65 is diabetes; 16 million individuals in the United States are diabetic and 40,000 per year suffer from ocular complications of the disease, often a result of retinal neovascularization.
  • laser photocoagulation has been effective in preventing severe visual loss in subgroups of high risk diabetic patients, the overall 10-year incidence of retinopathy remains substantially unchanged.
  • ARMD and diabetic retinopathy are the leading causes of visual loss in industrialized countries and do so as a result of abnormal retinal neovascularization. Since the retina consists of well-defined layers of neuronal, glial, and vascular elements, relatively small disturbances such as those seen in vascular proliferation or edema can lead to significant loss of visual function. Inherited retinal degenerations, such as retinitis pigmentosa (RP), are also associated with vascular abnormalities, such as arteriolar narrowing and vascular atrophy. While significant progress has been made in identifying factors that promote and inhibit angiogenesis, no treatment is currently available to specifically treat ocular vascular disease.
  • RP retinitis pigmentosa
  • Inherited degenerations of the retina affect as many as 1 in 3500 individuals and are characterized by progressive night blindness, visual field loss, optic nerve atrophy, arteriolar attenuation, altered vascular permeability and central loss of vision often progressing to complete blindness ( Heckenlively, J. R., editor, 1988; Retinitis Pigmentosa, Philadelphia: JB Lippincott Co. ). Molecular genetic analysis of these diseases has identified mutations in over 110 different genes accounting for only a relatively small percentage of the known affected individuals ( Humphries et al., 1992, Science 256:804-808 ; Farrar et al. 2002, EMBO J. 21:857-864 .).
  • Angiogenesis is the process by which new blood vessels form.
  • capillaries sprout from existing vessels, eventually growing in size as needed by the organism.
  • endothelial cells which line the blood vessels, divide in a direction orthogonal to the existing vessel, forming a solid sprout
  • Adjacent endothelial cells then form large vacuoles and the cells rearrange so that the vacuoles orient themselves end to end and eventually merge to form the lumen of a new capillary (tube formation).
  • Angiogenesis is stimulated by a number of conditions, such as in response to a wound, and accompanies virtually all tissue growth in vertebrate organisms such as mammals. Angiogenesis also plays a role in certain disease states such as certain cancers.
  • the growth of tumors for example, requires blood vessel growth to provide oxygen and nutrients to the growing tumor tissue.
  • ocular neovascularization is associated with the vast majority of eye diseases that lead to catastrophic loss of vision.
  • Angiogenesis may be arrested or inhibited by interfering with the chemical signals that stimulate the angiogenic process.
  • angiogenic endothelial cells produce proteases to digest the basal lamina that surround the blood vessels, thus clearing a path for the new capillary. Inhibition of these proteases, or their formation, can prevent new vessels from forming.
  • the endothelial cells proliferate in response to chemical signals.
  • Particularly important proliferation signals include the vascular endothelial growth factor (VEGF), and the fibroblast growth factor (FGF) families of proteins. VEGF has been shown to be involved in vascularization of certain tumors. Interference with these proliferation signaling processes can also inhibit angiogenesis.
  • VEGF vascular endothelial growth factor
  • FGF fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • angiogenesis In the normal adult, angiogenesis is tightly regulated, and is limited to wound healing, pregnancy and uterine cycling. Angiogenesis is turned on by specific angiogenic molecules such as basic and acidic fibroblast growth factor (FGF), VEGF, angiogenin, transforming growth factor (TGF), tumor necrosis factor- ⁇ (TNF- ⁇ ) and platelet derived growth factor (PDGF). Angiogenesis can be suppressed by inhibitory molecules such as interferon- ⁇ , thrombospondin-1, angiostatin and endostatin. It is the balance of these naturally occurring stimulators and inhibitors that controls the normally quiescent capillary vasculature. When this balance is upset, as in certain disease states, capillary endothelial cells are induced to proliferate, migrate and ultimately differentiate.
  • FGF basic and acidic fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • angiogenin transforming growth factor
  • TGF tumor necrosis factor- ⁇
  • PDGF platelet
  • Angiogenesis plays a central role in a variety of disease including cancer and ocular neovascularization. Sustained growth and metastasis of a variety of tumors has also been shown to be dependent on the growth of new host blood vessels into the tumor in response to tumor derived angiogenic factors. Proliferation of new blood vessels in response to a variety of stimuli occurs as the dominant finding in the majority of eye disease and that blind including proliferative diabetic retinopathy, ARMD, rubeotic glaucoma, interstitial keratitis and retinopathy of prematurity. In these diseases, tissue damage can stimulate release of angiogenic factors resulting in capillary proliferation.
  • VEGF plays a dominant role in iris neovascularization and neovascular retinopathies. While reports clearly show a correlation between intraocular VEGF levels and ischemic retinopathic ocular neovascularization, FGF likely plays a role as well. Basic and acidic FGF are known to be present in the normal adult retina, even though detectable levels are not consistently correlated with neovascularization. This may be largely due to the fact that FGF binds very tightly to charged components of the extracellular matrix and may not be readily available in a freely diffusible form that would be detected by standard assays of intraocular fluids.
  • a final common pathway in the angiogenic response involves integrin-mediated information exchange between a proliferating vascular endothelial cell and the extracellular matrix.
  • This class of adhesion receptors called integrins, are expressed as heterodimers having an ⁇ and P subunit on all cells.
  • integrins One such integrin, ⁇ v ⁇ 3 , is the most promiscuous member of this family and allows endothelial cells to interact with a wide variety of extracellular matrix components.
  • Peptide and antibody antagonists of this integrin inhibit angiogenesis by selectively inducing apoptosis of the proliferating vascular endothelial cells.
  • Two cytokine-dependent pathways of angiogenesis exist and may be defined by their dependency on distinct vascular cell integrins, ⁇ v ⁇ 3 , and ⁇ v ⁇ 5 .
  • basic FGF- and VEGF-induced angiogenesis depend on integrin ⁇ v ⁇ 3 and ⁇ v ⁇ 5 , respectively, since antibody antagonists of each integrin selectively block one of these angiogenic pathways in the rabbit corneal and chick chorioallantoic membrane (CAM) models.
  • Peptide antagonists that block all ⁇ , integrins inhibit FGF- and VEGF-stimulated angiogenesis.
  • ⁇ v ⁇ 3 and ⁇ v ⁇ 5 integrins are selectively displayed on blood vessels in tissues from patients with active neovascular eye disease. While only ⁇ v ⁇ 3 was consistently observed in tissue from patients with ARMD, ⁇ v ⁇ 3 and ⁇ v ⁇ 5 both were present in tissues from patients with proliferative diabetic retinopathy.
  • Systemically administered peptide antagonists of integrins blocked new blood vessel formation in a mouse model of retinal vasculogenesis.
  • anti-angiogenic agents have a role in treating retinal degeneration to prevent the damaging affects of these trophic and growth factors.
  • Angiogenic agents also have role in promoting desirable vascularization to retard retinal degeneration by enhancing blood flow to cells.
  • angiostatic therapies ultimately become successful.
  • Naturally occurring compensatory mechanisms may ultimately render angiogenic monotherapies obsolete.
  • Angiostatic drugs generally target a single cytokine or intracellular angiogenic pathway.
  • angiogenesis is likely to be initiated by the combined signaling of multiple pathways.
  • blocking a single pathway may be insufficient to prevent angiogenesis during the treatment of neovascular diseases.
  • it is also likely that blocking a single pathway induces compensation and increased roles of other angiogenic pathways.
  • WO 03/080648 relates to recombinant adeno-associated viral (rAAV) vector compositions comprising nucleic acid segments encoding therapeutic gene products, and their use in the manufacture of medicaments for treating various disorders of the eye including, for example, retinal, ocular or choroidal neovascularization (CNV).
  • rAAV adeno-associated viral
  • the present invention relates to compositions which an useful in a method of treating a neovascular disease, such as a retinal neovascular disease, by administering to a mammal suffering from a neovascular disease an amount of a combination of angiogenesis suppressing drugs sufficient to inhibit new blood vessel formation.
  • a neovascular disease such as a retinal neovascular disease
  • the present invention provides a composition
  • a composition comprising (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1 having the formula:
  • the angiostatic fragment of TrpRS preferably has the amino acid residue sequence of SEQ ID NO: 1.
  • composition may further comprise at least one therapeutic agent selected from the group consisting of an angiostatic steroid, an anti-neoplastic agent, an anti-bacterial agent, an anti-viral agent, and an anti-inflammatory agent.
  • angiostatic fragment ofTrpRS is a dimer.
  • composition of the first aspect of the present invention is for treating a neovascular disease.
  • the present invention provides the use of (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1 having the formula: for the preparation of a pharmaceutical composition for treatment of a neovascular disease in a mammal by administering a vascular development inhibiting amount of a combination of drugs comprising (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1.
  • the disease may be a retinal neovascular disease.
  • the combination of drugs is to be administered by intravitreal injection into the eye of a mammal suffering from a retinal neovascular disease, preferably selected from the group consisting of an ischemic retinopathy, a vascular hemorrhage, a vascular leakage, a choroidopathy, age related macular degeneration, diabetic retinopathy, presumed ocular histoplasmosis, retinopathy of prematurity, sickle cell anemia, and retinitis pigmentosa.
  • a retinal neovascular disease preferably selected from the group consisting of an ischemic retinopathy, a vascular hemorrhage, a vascular leakage, a choroidopathy, age related macular degeneration, diabetic retinopathy, presumed ocular histoplasmosis, retinopathy of prematurity, sickle cell anemia, and retinitis pigmentosa.
  • the present invention provides the use of (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1 having the formula: for the preparation of a pharmaceutical composition for treatment of a tumor in a mammal by administering a vascular development inhibiting amount of a combination of drugs comprising (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting ofSEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1.
  • the mammal is a human.
  • the angiostatic fragment of TrpRS has the amino acid residue sequence of SEQ ID NO: 1
  • the present invention provides a pharmaceutical composition comprising a composition of the first aspect of the present invention and a pharmaceutically acceptable carrier therefor.
  • Neovascular diseases treatable by the compositions of the present invention include, without limitation, ocular diseases such as retinal degenerative diseases, retinal vascular degenerative diseases, ischemic retinopathies, vascular hemorrhages, vascular leakage, and choroidopathies in neonatal, juvenile, or fully mature mammals.
  • the compositions of the present invention can also be utilized to treat neovascular diseases such as solid tumor cancers (e.g., lung cancer, breast cancer, and prostate cancer) and rheumatoid arthritis, for example.
  • the angiostatic fragments of TrpRS are a 43 kDa fragment (i.e., the T2 fragment, "T2-TrpRS”, SEQ ID NO: 1, or a mutant of T2-TrpRS, "T2-TrpRS-GD", SBQ ID NO: 2; both of which are shown in Figure 1 ), the 48 kDa fragment known as mini-TrpRS (SBQ ID NO: 3, shown in Figure 2 ), and the 46 kDa fragment known as T1-TrpRS (SEQ ID NO: 4, shown in Figure 2 ).
  • the amino acid residue sequence of T2-TrpRS-GD (SEQ ID NO: 2), differs from SEQ ID NO: 1 by two amino acid residue substitutions (i.e., S121G and Y122D).
  • the amino acid residue sequence of full length human TrpRS (SEQ ID NO: 5) is shown in Figure 3 , along with an indication of the position of the T1, T2 and mini fragments thereof. Without being bound by theory, it is believed that the angiostatic fragments of TrpRS can form noncovalent dimers (see e.g., Yu et al. J. Biol. Chem. 2004, 279: 8378-8388 ), which may contribute to the biological activity of the fragments.
  • TrpRS angiostatic fragment of TrpRS
  • T1-TRpRS e.g., T1-TRpRS, T2-TrpRS, mini-TrpRS
  • the compound having the formula of Compound (1) is available from Merck KGaA (Darmstadt, Germany) as EMD 472523 and is a peptidomimetic integrin signaling inhibitor.
  • the VEGF signaling inhibitor pegaptanib sodium (Compound (2)) is a nuclease resistant, 2'-fluoropyrimidine RNA-based aptamer specific for VEGF-165 and is a polyethoxylated oligonucleotide having the following formula (SEQ ID NO: 6; Figure 59 , R in Figure 59 is a 40 kiloDalton polyethylene glycol (PEG) chain): 5'-40K PEG-C5 aminolinker-CfGmGmArArUfCfAmGmUfGmAmAmUfGmCfUf UfAmUfAmCfAmUfCfCfCm3' - 3'dT wherein
  • a polyethoxylated oligonucleotide of SEQ ID NO: 6 is sold commercially under the trademark MACUGEN® by from Eyetech Pharmaceuticals, Inc., and is also known as NX1838 or pegaptanib sodium.
  • the combination of drugs also includes at least one additional therapeutic agent such as an angiostatic steroid, an anti-neoplastic agent, an anti-bacterial agent, an anti-viral agent, an anti-inflammatory agent, and the like.
  • angiostatic steroid such as an angiostatic steroid, an anti-neoplastic agent, an anti-bacterial agent, an anti-viral agent, an anti-inflammatory agent, and the like.
  • angiostatic steroids examples include anecortave acetate and triamcinolone acetonide.
  • anti-neoplastic agents include Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthranaycin; Asparaginase; Asperlin ; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer, Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefingol; Chlorambucil; Cirolemycia ; Cisplatin; Cladrib
  • Suitable anti-bacterial agents include, but are not limited to, penicillins, aminoglycosides, macrolides, monobactams, rifamycins, tetracyclines, chloramphenicol, clindamycin, lincomycin, imipenem, fusidic acid, novobiocin, fosfomycin, fusidate sodium, neomycin, polymyxin, capreomycin, colistimethate, colistin, gramicidin, minocycline, doxycycline, vanomycin, bacitracin, kanamycin, gentamycin, erythromicin and cephalosporins.
  • Suitable anti-inflammatory agents include, but are not limited to, aspirin (acetylsalicylic acid), indomethacin, sodium indomethacin trihydrate, salicylamide, naproxen, colchicine, fenoprofen, sulindac, diflunisal, diclofenac, indoprofen and sodium salicylamide.
  • anti-viral agents include, but are not limited to, alpha-methyl-P-adamantane methylamine, 1-D-ribofuranosyl-1,2,4-triazole-3 carboxamide, 9-(2 hydroxy-ethoxy)methylguavine, adamaatanamine, 5-iodo-2'-deoxyuiidine, trifluorothymidine, interferon, adenine arabinoside, CD4, 3'-azido-3'-deoxythymidine (AZT), 9-(2-hydroxyethoxymethyl)-guanine (acyclovir), phosphonofomic acid, 1-adamaatanamine, peptide T, and 2',3'-dideoxycytidine.
  • alpha-methyl-P-adamantane methylamine 1-D-ribofuranosyl-1,2,4-triazole-3 carboxamide
  • 9-(2 hydroxy-ethoxy)methylguavine adamaatanamine
  • Neovascular diseases treatable by the compositions of the present invention include, without limitation, neovascular diseases of the eye (e.g., retinal and choroidal neovascular diseases), rubeotic glaucoma, pterygia, solid tumor cancers (e.g., lung cancer, breast cancer, and prostate cancer), osteoarthritis, rheumatoid arthritis, vascular anomalies and malformations (e.g., hemangiomas, lymphangiomas, and the line), and psoriasis.
  • neovascular diseases of the eye e.g., retinal and choroidal neovascular diseases
  • rubeotic glaucoma e.g., rubeotic glaucoma
  • pterygia solid tumor cancers (e.g., lung cancer, breast cancer, and prostate cancer)
  • osteoarthritis e.g., rheumatoid arthritis
  • vascular anomalies and malformations e.g
  • compositions of the present invention may be used in a method of treating retinal neovascular diseases in a mammal.
  • the method comprises intravitreally injecting into the eye of a mammal suffering from a neovascular disease a vascular development inhibiting amount of a combination of antiangiogenic and angiostatic compositions that provide an angiostatic fragment of TrpRS as described above, the VEGF signaling inhibitor pegaptanib sodium; and the integrin signaling inhibitor compound (1).
  • This method can be utilized to treat ocular diseases such as vascular degenerative diseases, ischemic retinopathies, vascular hemorrhages, vascular leakage, and choroidopathies in neonatal, juvenile or fully mature mammals.
  • ocular diseases such as vascular degenerative diseases, ischemic retinopathies, vascular hemorrhages, vascular leakage, and choroidopathies in neonatal, juvenile or fully mature mammals.
  • diseases include age related macular degeneration, diabetic retinopathy, presumed ocular histoplasmosis, retinopathy of prematurity, sickle cell anemia, hemangioma, pterygia, ischemic central retinal vein occlusion, blanch retinal vein occlusion, ocular melanoma, retinal blastoma, and retinitis pigmentosa, as well as retinal injuries.
  • Another aspect of the present invention is a therapeutic composition useful for the treatment of neovascular diseases, which comprises an angiostatic fragment of TrpRS as described above, the VEGF signaling inhibitor pegaptanib sodium, and the integrin signaling inhibitor compound (1), together with one or more pharmaceutically acceptable excipient.
  • the composition further comprises at least one additional therapeutic agent such as an angiostatic steroid, an anti-neoplastic agent, an anti-bactenal agent, an anti-viral agent, an anti-inflammatory agent, and the like.
  • at least one additional therapeutic agent such as an angiostatic steroid, an anti-neoplastic agent, an anti-bactenal agent, an anti-viral agent, an anti-inflammatory agent, and the like.
  • compositions of the present invention may be used in a method of treating a neovascular disease comprising administering to a mammal suffering from a neovascular disease a vascular development inhibiting amount of a combination of drugs comprising an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) as described above, the vascular endothelial growth factor (VEGF) signaling inhibitor pegaptanib sodium and the integrin signaling inhibitor compound (1).
  • TrpRS angiostatic fragment of tryptophanyl-tRNA synthetase
  • VEGF vascular endothelial growth factor
  • a vascular development inhibiting amount of a composition of the present invention is at least about 10 ⁇ g/kg body weight and, in most cases, not in excess of about 8 mg/kg body weight per day for systemic treatments.
  • the dosage is in the range of about 10 ⁇ g/kg body weight to about 1 mg/kg body weight daily.
  • the preferred dosage is in the range of about 0.1 to about 5 milligrams per eye for a given treatment.
  • the compositions may be administered in a single dose or in multiple doses over time.
  • One of ordinary skill in the medical arts would be capable of determining the optimum effective therapeutic dosage of a composition of the present invention, taking into account the particular patient, the drugs present in the composition, the disease state, and other factors that are well known in the medical arts.
  • compositions of this invention can be embodied in a variety of physical forms. These forms include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspensions, aerosols, liposomes, suppositories, injectable and infusible solutions and sustained release forms.
  • solid, semi-solid, and liquid dosage forms such as tablets, pills, powders, liquid solutions or suspensions, aerosols, liposomes, suppositories, injectable and infusible solutions and sustained release forms.
  • a therapeutic composition according to this invention can be administered by conventional routes of administration, such as parenteral, subcutaneous, intravenous, intramuscular, intralesional, intrasternal, intravitreal, intracranial, or aerosol routes.
  • Topical routes of administration can also be used, with application of the compositions locally to a particular part of the body (e.g., eye, skin, lower intestinal tract, vagina, rectum) where appropriate.
  • the therapeutic compositions also include conventional pharmaceutically acceptable carriers and excipients that are known to those of skill in the art.
  • compositions of the present invention can be formulated and administered using methods and compositions similar to those used for the individual classes of active ingredients present in the compositions. It will be understood by those of skill in the art that conventional doses will vary depending upon the particular active ingredients in the composition, as well as the patient's health, weight, age, sex, the condition or disease, and the desired mode of administration.
  • compositions of this invention include pharmacologically appropriate, pharmaceutically acceptable carriers, excipients and vehicles.
  • these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media such as phosphate buffered saline (PBS).
  • Parenteral vehicles can include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
  • intravenous vehicles can include fluid and nutrient replenishers, and electrolyte replenishers, such as those based on Ringer's dextrose.
  • Excipients such as preservatives and other additives can also be present, such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases.
  • Suitable formulation aids, carriers, other excipients, and methods of formulating pharmaceutical compositions are disclosed in Remington's Pharmaceutical Sciences, 14th Ed., Mack Publishing Co., 1970, particularly Part VIII, "pharmaceutical Preparations and Their Manufacture", pages 1461 - 1762 .
  • compositions of the present invention can be packaged in suitably sterilized bottles or vials, either in multi-dose or in unit dose forms.
  • the containers are preferably hermetically sealed after being filled with a composition of the invention.
  • the compositions are packaged in a container having a label affixed thereto, which label identifies the drugs present in the composition, and bears a notice in a form prescribed by a government agency such as the United States Food and Drug Administration, reflecting approval of the composition under appropriate laws, dosage information, and the like.
  • the label preferably contains information about the composition that is useful to a health care professional administering the composition to a patient.
  • the package also preferably contains printed informational materials relating to the administration of the composition, instructions, indications, and any necessary required warnings.
  • the second phase of retinal vessel formation begins between postnatal days 7 (P7) and 10 (P10) when collateral branches sprout from capillaries of the superficial plexus and penetrate into the retina where their tips branch and anastamose laterally to form a planar "deep vascular plexus". While the deep vascular plexus is in place by P14, it undergoes extensive remodeling from P14 to P21. It is of interest to note that the formation of these vascular networks in the neonatal mouse are strikingly similar to the events occurring in the third trimester human fetus.
  • the reproducibility of the murine retinal development process and its easy accessibility in neonatal animals provide an opportunity to assess the efficacy of antiangiogenic compounds in a physiologically relevant model of angiogenesis. Additional advantages of the neonatal mouse model are the ability to qualitatively and quantitatively evaluate the angiostatic effect of putative antagonists of angiogenesis.
  • Angiostatic activity was evaluated based upon the degree of angiogenesis in the deep, outer retinal vascular layer (secondary layer) that develops between P8 and P12.
  • the appearance of the inner blood vessel network (primary layer) was evaluated for normal development and signs of toxicity. No abnormalities in the inner vascular layer were observed in any of the assays performed and described herein.
  • Qualitative evaluation of the secondary layer vascularization can be performed by microscopically photographing appropriately stained superficial and deep layers of excised retinas and determining the percentage of eyes in which formation of the deep vascular layer is completely or partially inhibited. All data presented herein are based on qualitative analysis of the percentage of eyes that demonstrated a 75 to 100 % inhibition of deep retinal vascular network formation after treatment. In most cases, the percentage of mice that exhibited > 95 % and 100 % inhibition of deep retinal vascular network formation are also provided.
  • Peptidomimetic integrin signaling inhibitor Compound (1) is solubilized in PBS at a concentration of about 20 mg/ml in PBS (1x concentration).
  • T2-TrpRS is solubilized in PBS at a concentration of about 0.5 mg/ml (1x concentration).
  • VEGF aptamer pegaptanib sodium; Compound (2) is solubilized in PBS at a concentration of about 2 mg/ml (1x concentration) to achieve an injection of approximately 1 ⁇ g/eye at 1x concentration.
  • the compounds were prepared at 2 or 3 times the 1x concentration of each material and then combined to produce a final solution containing each individual compound at the same concentration as was used alone (e.g., 1x, 0.5x, 0.25x or 0.1x, as the case may be).
  • a single injection of 0.5 ⁇ l of PBS solutions of the drugs was administered intravitreally regardless of the number of compounds being injected.
  • the term "0.1x” refers to one tenth of the 1x concentration of a given material
  • 0.25x refers to one quarter of the 1x concentration of a given material
  • 0.5x refers to one half of the 1x concentration of a given material, and so forth for similar designations.
  • OIR Oxygen Induced Retinopathy
  • Quantification of neovascularization in the OIR model involved quantification of neovascular tuft formation, as well as quantification of obliteration.
  • Retinal whole mounts are prepared and blood vessels thereof are stained with isolectin GS-IB 4 .
  • Confocal imaging focusing just above the superficial vascular plexus, is carried out and a montage of four quadrants is made.
  • Neovascular tufts are identified (Adobe PHOTOSHOP®) and the area of pixelation is quantified
  • areas of obliteration are traced (Adobe PHOTOSHOP®), and the area of pixelation is quantified.
  • Conversion factor based on image acquisition (resolution, size etc.) is then applied to obtain a value in ⁇ m 2 .
  • An in vivo angiogenesis assay in the neonatal mouse (Balb/C, The Jackson Laboratory, Bar Harbor, ME) was used to evaluate the angiostatic activity of integrin signaling inhibitor Compound (1), T2-TrpRS, and VEGF aptamer Compound (2).
  • Intravitreous injection and retina isolation was performed with a dissecting microscope (SMZ 645, Nikon, Japan).
  • An eyelid fissure was created at postnatal day 7 (P7) with a fine blade to expose the globe for injection.
  • the samples (0.5 ⁇ l) were injected with a Hamilton syringe fitted with a 32-gauge needle (Hamilton Company, Reno, NV). The injection was made between the equator and the corneal limbus. During injection, the location of the needle tip was monitored by direct visualization to determine that it was in the vitreal cavity. Eyes with needle-induced lens or retinal damage were excluded from the study. After the injection, the eyelids were repositioned to close the fissure.
  • the blood vessels were specifically visualized by staining the retina for about 18 hours at about 4 °C with a rabbit antimouse collagen IV antibody (Chemicon, Temecula, CA) diluted 1:200 in blocking buffer or with a fluorescent conjugated isolectin ( Griffonia simplicifolia , Molecular Probes).
  • a rabbit antimouse collagen IV antibody (Chemicon, Temecula, CA) diluted 1:200 in blocking buffer or with a fluorescent conjugated isolectin ( Griffonia simplicifolia , Molecular Probes).
  • An ALEXA FLUOR® Alexa 594-conjngated goat anti-rabbit IgG antibody (Molecular Probes, Eugene, OR) (1:200 dilution in blocking buffer) was incubated with the retina for about 2 hours at about 4°C.
  • the retinas were then mounted for microscopic evaluation with slow-fade mounting media (Molecular Probes, Eugene, OR).
  • Example 1 Treatment of neonatal mouse eyes with a combination of a peptidomimetic integrin signaling inhibitor and a VEGF signaling inhibitor.
  • mice were intravitreally injected on postnatal day 8 (P8) with either a 0.25x concentration of integrin signaling inhibitor Compound (1) (five mice), a 0.5x concentration of VEGF aptamer Compound (2) (five mice), or a combination of a 0.25x concentration Compound (1) and a 0.5x concentration of Compound (2) (six mice).
  • P8 postnatal day 8
  • another group of six mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 1 and in Figures 4 and 5 .
  • Example 2 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 0.1x concentration of T2-TrpRS (eight mice), a 0.1x concentration of VEGF aptamer Compound (2) (eight mice), or a combination of a 0.1x concentration T2-TrpRS and a 0.1x concentration of Compound (2) (ten mice).
  • T2-TrpRS 0.1x concentration of T2-TrpRS
  • VEGF aptamer Compound (2) a combination of a 0.1x concentration T2-TrpRS and a 0.1x concentration of Compound (2)
  • PBS intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 2 and in Figures 6 , 7 , 8 , and 9 .
  • Example 3 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS (eight mice), a 1x concentration of VEGF aptamer Compound (2) (eight mice), or a combination of a 1x concentration T2-TrpRS and a 1x concentration of Compound (2) (ten mice).
  • T2-TrpRS a 1x concentration of T2-TrpRS
  • VEGF aptamer Compound (2) a 1x concentration of Compound (2)
  • Compound (2) ten mice.
  • ten mice ten mice.
  • another group of six mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes.
  • Example 4 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS (eight mice), a 0.5x concentration of VEGF aptamer Compound (2) (ten mice) or a combination of a 1x concentration T2-TrpRS and a 0.5x concentration of Compound (2) (ten mice).
  • T2-TrpRS epidermal growth factor
  • T2-TrpRS vascular endothelial growth factor aptamer Compound (2)
  • Compound (2) ten mice.
  • Another group of six mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes.
  • Example 5 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and an integrin signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 0.5x concentration of integrin signaling inhibitor Compound (1) or a combination of a 1x concentration T2-TrpRS and a 0.5x concentration of Compound (1), in groups of six mice for each treatment regimen.
  • a control another group of four mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes.
  • Example 6 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 0.5x concentration of integrin signaling inhibitor Compound (1), a 1x concentration of VEGF aptamer Compound (2), a combination of a 1x concentration T2-TrpRS and a 0.5x concentration of Compound (1), a combination of a 1x concentration T2-TrpRS and a 1x concentration of Compound (2), or a combination of a 1x concentration T2-TrpRS, a 0.5x concentration of Compound (1), and a 1x concentration of Compound (2), in groups of eight mice for each treatment regimen.
  • mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 6 and in Figures 22 , 23 , 24 , 25 , 26 , 27 , and 28 .
  • Table 6 % Inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100% PBS 100 0 0 0 0 0 0 0.5x Comp.
  • Example 7 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 0.5x concentration of integrin signaling inhibitor Compound (1), a 1x concentration of VEGF aptamer Compound (2), a combination of a 0.5x concentration Compound (1) and a 1x concentration of Compound (2), or a combination of a 1x concentration T2-TrpRS, a 0.5x concentration of Compound (1), and a 1x concentration of Compound (2), in groups of eight mice for each treatment regimen.
  • a 1x concentration of T2-TrpRS a 0.5x concentration of Compound (1)
  • a 1x concentration of Compound (2) in groups of eight mice for each treatment regimen.
  • another group of six mice received only an intravitreal injection of PBS.
  • mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 7 and in Figures 29 , 30 , 31 , 32 , 33 , and 34 .
  • Example 8 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS (ten mice), a 0.25x concentration of VEGF aptamer Compound (2) (eleven mice), or a combination of a 1x concentration T2-TrpRS and a 0.25x concentration of Compound (2) (eleven mice).
  • a 1x concentration of T2-TrpRS ten mice
  • a 0.25x concentration of VEGF aptamer Compound (2) a combination of a 1x concentration T2-TrpRS and a 0.25x concentration of Compound (2)
  • PBS intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 8.
  • Example 9 Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor.
  • mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 1x concentration of integrin signaling inhibitor Compound (1), a 1x concentration of VEGF aptamer Compound (2), a combination of a 1x concentration T2-TrpRS and a 1x concentration of Compound (1), a combination of a 1x concentration Compound (1) and a 1x concentration of Compound (2), or a combination of a 1x concentration T2-TrpRS, a 1x concentration of Compound (2), and a 1x concentration of Compound (2).
  • another group of eight mice received only an intravitreal injection of PBS.
  • mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 9 and Figures 35-42 .
  • Example 10 Treatment of neonatal mouse eyes with varying doses of an angiostatic fragment of TrpRS.
  • mice were intravitreally injected on P4 with T2-TrpRS, at concentrations of 0.1x (8 mice), 0.3x (12 mice), 1x (12 mice), 2x (12 mice), and 3x (12 mice).
  • T2-TrpRS concentrations of 0.1x
  • mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 10. Table 10.
  • Example 11 Treatment of neonatal mouse eyes with varying doses of an angiostatic fragment of TrpRS.
  • mice were intravitreally injected on P4 with T2-TrpRS, at concentrations of 0.3x (6 mice), 1x (14 mice), 2x (8 mice), 3x (7 mice), and 5x (6 mice).
  • T2-TrpRS concentrations of 0.3x
  • 1x 14 mice
  • 2x 8 mice
  • 3x 7 mice
  • 5x 6 mice
  • another group of 10 mice received only an intravitreal injection of PBS.
  • the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure.
  • the vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 11. Table 11.
  • compositions comprising at least two materials selected from the group consisting of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor afford unexpectedly greater efficacy for inhibition of neovascularization in the neonatal mouse eye model than the expected levels of inhibition from the simple additive effects of the combination of individual components.
  • Table 12 clearly shows an unexpectedly higher level of inhibition of blood vessel formation in the deep vascular layer for mouse eyes treated with the compositions comprising combinations of at least two of Compound (1), Compound (2) and T2-TrpRS compared to the inhibition levels of the treatments with the individual inhibitors by themselves or the numerical sum thereof.
  • Table 12. Composite Data from the Examples. %Inhibition: 0-10% 16-15% 15-50% 50-75% 75-100% >95% 100% PBS (38 mice) 84.2 10.5 53 0 0 0 0 1x-2x Comp. (1) (30 mice) 33.3 20 133 16.7 16.7 0 0 1x Comp.
  • mice 39.1 20 7.6 11.9 21.4 4.7 2.4 1x 77-TrpRS (46 mice) 23.9 17.4 15.2 8.7 34.8 8.7 2.2
  • T2-TrpRS Tryptophan tRNA synthetase
  • mice are born without a retinal vasculature. During the first three weeks after birth an adult-like vasculature develops. The retinal vasculature forms three distinct planar plexuses with the superficial vascular plexus developing during the first post-natal week. At post-natal day 8 (P8), the vessels of the superficial plexus branch and migrate toward the deep plexus at the outer edge of the inner nuclear layer.
  • ⁇ v ⁇ 3 and ⁇ v ⁇ 5 integrin antagonist (Compound (1)) was stored as lyophilized powder in a desiccator at room temperature (I.A.) or -20°C (V.A.), and solubilized in sterile, RNAse-free 1x PBS immediately before use.
  • the VEGF aptamer (Compound (1)) was synthesized as a 40 kDa PEG conjugated compound (Transgenomic Inc, Boulder, CO) based on published information, Bridonneau, et al., J. Chromatogr: B. Diomed. Sci. App. 726: 237-47 (1999 ). The compound was determined to be pure by reverse phase liquid chromatography.
  • Concentrations reported herein refer to the final concentration of the active VEGF aptamer rather than the total concentration of the PEG conjugated compound and were determined by spectrophotometric analysis at 260/280 nm.
  • T2-TrpRS peptide was made as a recombinant compound as described in Otani, et al., Proc. Nat'l. Acad. Sci., USA 99: 178-183 (2002 ) and U.S. Provisional Application for Patent Serial No. 60/598,019 filed August 2, 2004 .
  • Purified product was stored in 50% glycerol at -20 °C, and dialyzed into sterile 1x PBS immediately before use.
  • Combination solutions were prepared by initially creating 3x stock solutions of each individual compound. The compounds were then mixed together, and with PBS where appropriate, to make a final solution containing each desired compound at a concentration equivalent to each corresponding monotherapy concentration.
  • Intravitreal injections All animal work adhered to strict protocol guidelines for the humane care and use of animals. Intravitreal injections were performed, the retinas were dissected, and the vasculature was visualized. OIR was induced according to the protocol described by Smith, et al., Invest. Ophthalmol. Vis. Sci., 35: 101-111 (1994 ), by exposing post-natal day 7 (P7) pups and their mothers to an environment of 75% oxygen (hyperoxia) for 5 days, followed by a return to room air (normoxia). Intravitreal injections were performed at P12, immediately following return to normoxia and the retinas were analyzed at P 17.
  • Blood vessels were stained using isolectin GS-IB 4 from Griffonia simplicifolia (lectin GS), conjugated to Alexa Fluor 594 (Molecular Probes, 1:150 dilution in PBS).
  • Confocal images were taken using a 4x objective lens, carefully focusing just above the inner limiting membrane of the retina, making the pre-laminar neovascular tufts prominent.
  • Four overlapping images were acquired from each retina and each individual image was converted to a 2 x 2 inch size with 300 pixels per inch.
  • the neovascular tuft areas were quantified by masked individuals using retinal whole mounts.
  • Tufts were specifically selected, based on their characteristic appearance and the higher intensity of isolectin staining, using the magic wand tool in Adobe PHOTOSHOP® software. The total area in pixels was then determined. The areas of neovascular tuft formation were normalized to PBS-injected control OIR retinas.
  • Dosing Dosing experiments were first performed to determine the maximum efficacy dose for each individual compound. Each compound was found to have a bell-shaped efficacy curve with optimal effective doses of 5-10 ⁇ g (10-20 nmoles) per eye for the integrin antagonist ( Figure 46 ), 1.0-2.0 ⁇ g (108-215 pmoles) per eye for the VEGF aptamer ( Figure 44 ), and 0.25-0.5 ⁇ g (5.2-10.4 pmoles) per eye for T2-TrpRS ( Figure 43 ). Single injections of each monotherapy at the optimal dose, and solutions containing appropriate combinations of each compound at equivalent doses were then performed to compare the aagiostatic activities.
  • Neonatal mouse angiogenesis model combination experiment Percentage of retinas with the indicated levels of neovascular inhibition Injection N 0-10% 10-25% 25-50% 50-75% >75% >90% 100% PBS 38 84.2 10.5 5.3 0.0 0.0 0.0 0.0 5-10 ⁇ g integrin antagonist 30 33.3 20.0 13.3 16.7 16.7 0.0 0.0 1-2 ⁇ g VEGF aptamer 42 39.1 20.0 7.6 11.9 21.4 4.7 2.4 0.25 ⁇ g T2-TtpRS 46 23.9 17.4 15.2 8.7 34.8 8.7 2.2 77-TrpRS + integrin ant. 22 18.2 13.6 9.1 9.1 50.0 27.3 13.6 Integrin ant. + VEGF apt.
  • the mouse model of oxygen-induced retinopathy (OIR) described hereinabove is a well-accepted model of hypoxia-induced neovascularization in the retina.
  • the associated vascular changes are consistent, reproducible and quantifiable.
  • the use of this model has been extended to the general study of disease-related ischemic vasculopathies and related anti-angiogenic interventions.
  • combination therapies demonstrated improved angiostatic activities compared to the monotherapies.
  • each monotherapy and the various combination therapies were tested at one-tenth of the optimal doses.
  • the concentration of each compound in the combination solutions was equivalent to the corresponding monotherapy concentration.
  • no significant inhibition of pathological neovascular tuft formation was observed following monotherapy treatments.
  • significant reductions in neovascular tuft formations were observed using each double combination ( Figure 57 ).
  • At least two anti-angiogenic therapies are combined (e.g., a combination of VEGF signaling inhibitor, such as a VEGF aptamer, combined with an angiostatic fragment of TrpRS, such as the T2 fragment of TrpRS, and optionally and integrin antagonist).
  • VEGF signaling inhibitor such as a VEGF aptamer
  • TrpRS angiostatic fragment of TrpRS
  • compositions of the present invention comprising an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS), a vascular endothelial growth factor (VBGF) signaling inhibitor, and an integrin signaling inhibitor, provide a new and surprisingly efficacious treatment regimen for neovascular diseases, particularly for neovascular diseases of the eye.
  • TrpRS tryptophanyl-tRNA synthetase
  • VBGF vascular endothelial growth factor
  • Glioblastoma multiform is an incurable malignant brain tumor, usually fatal within one year of diagnosis.
  • the 9L rat gliosarcoma cell line is used as a model for malignant gliomas.
  • the tumor is highly vascularized and infiltrates into normal brain tissue.
  • Untreated animal receiving a bolus of 9L cells intracerebrally have a survival tie of approximately 3 weeks, once the tumor cells have been implanted.
  • intracerebral 9L tumors were established by stereotactic inoculation of about 50,000 cells in about 2 ⁇ l of Dulbecco's Modified Eagles's Medium (DMEM; Life Technologies, Gaithersburg, MD) into the right frontal lobe of CD 344 Fisher rats that had previously been anesthetized with ketamine and xylazine.
  • DMEM Dulbecco's Modified Eagles's Medium
  • a 10 ⁇ l bolus of a composition of the invention (4.5 mg/l of 17-TrpRS, 30 mg/l of Compound (1), and 6 mg/l of Compound (2); pegaptanib sodium) was injected stereotactically over the course of about 2 minutes into the same region of the brain as the 9L cells were implanted.
  • a pump was inserted into a subcutaneous pocket between the scapulae.
  • a catheter connected via tubing to the pump was inserted into the same burr hole made for introduction of the 9L cells, and was fixed in place.
  • Each pump had a flow rate of about 8 ⁇ l per hour.
  • An additional quantity of the composition of the invention was continuously pumped into the brain of each animal for about 24 hours. The continuous pumping distributed the composition throughout the entire hemisphere of the brain in which the tumor cells had been implanted.
  • Nine rats received the composition of the invention and nine rats received straight PBS as a control group.

Abstract

The present invention provides compositions and methods of treating neovascular diseases, such as a retinal neovascular diseases and tumors, by administering to a patient suffering from a neovascular disease or tumor a vascular development inhibiting amount of a combination of the angiogenesis suppressing drugs comprising an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) and at least one compound selected from the group consisting of a vascular endothelial growth factor (VEGF) signaling inhibitor and an integrin signaling inhibitor. Compositions for use in the methods include an admixture of an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) and at least one of a vascular endothelial growth factor (VEGF) signaling inhibitor and an integrin signaling inhibitor, together with a pharmaceutically acceptable excipient.

Description

    Field of the Invention
  • This invention relates to compositions comprising a combination of angiostatic and antiangiogenic drugs said compositions being useful in the treatment of neovascular diseases, such as retinal neovascular diseases.
  • Background of the Invention
  • The vast majority of diseases that cause catastrophic loss of vision do so as a result of ocular neovascularization. For example, age related macular degeneration (ARMD) affects 12-15 million Americans over the age of 65 and causes visual loss in 10-15% of them as a direct effects of choroidal (sub-retinal) neovascularization. The leading cause of visual loss for Americans under the age of 65 is diabetes; 16 million individuals in the United States are diabetic and 40,000 per year suffer from ocular complications of the disease, often a result of retinal neovascularization. While laser photocoagulation has been effective in preventing severe visual loss in subgroups of high risk diabetic patients, the overall 10-year incidence of retinopathy remains substantially unchanged. For patients with choroidal neovascularization due to ARMD or inflammatory eye disease such as ocular histoplasmosis, photocoagulation, with few exceptions, is ineffective in preventing visual loss. While recently developed, non-destructive photodynamic therapies hold promise for temporarily reducing individual loss in patients with previously untreatable choroidal neovascularization, only 61.4% of patients treated every 3-4 months had improved or stabilized vision compared to 45.9% of the placebo-treated group.
  • ARMD and diabetic retinopathy are the leading causes of visual loss in industrialized nations and do so as a result of abnormal retinal neovascularization. Since the retina consists of well-defined layers of neuronal, glial, and vascular elements, relatively small disturbances such as those seen in vascular proliferation or edema can lead to significant loss of visual function. Inherited retinal degenerations, such as retinitis pigmentosa (RP), are also associated with vascular abnormalities, such as arteriolar narrowing and vascular atrophy. While significant progress has been made in identifying factors that promote and inhibit angiogenesis, no treatment is currently available to specifically treat ocular vascular disease.
  • Inherited degenerations of the retina affect as many as 1 in 3500 individuals and are characterized by progressive night blindness, visual field loss, optic nerve atrophy, arteriolar attenuation, altered vascular permeability and central loss of vision often progressing to complete blindness (Heckenlively, J. R., editor, 1988; Retinitis Pigmentosa, Philadelphia: JB Lippincott Co.). Molecular genetic analysis of these diseases has identified mutations in over 110 different genes accounting for only a relatively small percentage of the known affected individuals (Humphries et al., 1992, Science 256:804-808; Farrar et al. 2002, EMBO J. 21:857-864.). Many of these mutations are associated with enzymatic and structural components of the phototransduction machinery including rhodopsin, cGMP phosphodiesterase, rds peripherin, and RPE65. Despite these observations, there are still no effective treatments to slow or reverse the progression of these retinal degenerative diseases. Recent advances in gene therapy have led to successful reversal of the rds (Ali et al. 2000, Nat. Genet. 25:306-310) and rd (Takahashi et al. 1999, J. Virol. 73:7812-7816) phenotypes in mice and the RPE65 phenotype in dogs (Acland et al. 2001, Nat. Genet. 28:92-95) when the wild type transgene is delivered to photoreceptors or the retinal pigmented epithelium (RPE) in animals with a specific mutation.
  • Angiogenesis is the process by which new blood vessels form. In response to specific chemical signals, capillaries sprout from existing vessels, eventually growing in size as needed by the organism. Initially, endothelial cells, which line the blood vessels, divide in a direction orthogonal to the existing vessel, forming a solid sprout Adjacent endothelial cells then form large vacuoles and the cells rearrange so that the vacuoles orient themselves end to end and eventually merge to form the lumen of a new capillary (tube formation).
  • Angiogenesis is stimulated by a number of conditions, such as in response to a wound, and accompanies virtually all tissue growth in vertebrate organisms such as mammals. Angiogenesis also plays a role in certain disease states such as certain cancers. The growth of tumors, for example, requires blood vessel growth to provide oxygen and nutrients to the growing tumor tissue. In addition, ocular neovascularization is associated with the vast majority of eye diseases that lead to catastrophic loss of vision.
  • Angiogenesis may be arrested or inhibited by interfering with the chemical signals that stimulate the angiogenic process. For example, angiogenic endothelial cells produce proteases to digest the basal lamina that surround the blood vessels, thus clearing a path for the new capillary. Inhibition of these proteases, or their formation, can prevent new vessels from forming. Likewise, the endothelial cells proliferate in response to chemical signals. Particularly important proliferation signals include the vascular endothelial growth factor (VEGF), and the fibroblast growth factor (FGF) families of proteins. VEGF has been shown to be involved in vascularization of certain tumors. Interference with these proliferation signaling processes can also inhibit angiogenesis.
  • Several factors are involved in angiogenesis. Both acidic and basic fibroblast growth factor molecules are mitogens for endothelial cells and other cell types. A highly selective mitogen for vascular endothelial cells is VEGF.
  • In the normal adult, angiogenesis is tightly regulated, and is limited to wound healing, pregnancy and uterine cycling. Angiogenesis is turned on by specific angiogenic molecules such as basic and acidic fibroblast growth factor (FGF), VEGF, angiogenin, transforming growth factor (TGF), tumor necrosis factor-α (TNF-α) and platelet derived growth factor (PDGF). Angiogenesis can be suppressed by inhibitory molecules such as interferon-α, thrombospondin-1, angiostatin and endostatin. It is the balance of these naturally occurring stimulators and inhibitors that controls the normally quiescent capillary vasculature. When this balance is upset, as in certain disease states, capillary endothelial cells are induced to proliferate, migrate and ultimately differentiate.
  • Angiogenesis plays a central role in a variety of disease including cancer and ocular neovascularization. Sustained growth and metastasis of a variety of tumors has also been shown to be dependent on the growth of new host blood vessels into the tumor in response to tumor derived angiogenic factors. Proliferation of new blood vessels in response to a variety of stimuli occurs as the dominant finding in the majority of eye disease and that blind including proliferative diabetic retinopathy, ARMD, rubeotic glaucoma, interstitial keratitis and retinopathy of prematurity. In these diseases, tissue damage can stimulate release of angiogenic factors resulting in capillary proliferation. VEGF plays a dominant role in iris neovascularization and neovascular retinopathies. While reports clearly show a correlation between intraocular VEGF levels and ischemic retinopathic ocular neovascularization, FGF likely plays a role as well. Basic and acidic FGF are known to be present in the normal adult retina, even though detectable levels are not consistently correlated with neovascularization. This may be largely due to the fact that FGF binds very tightly to charged components of the extracellular matrix and may not be readily available in a freely diffusible form that would be detected by standard assays of intraocular fluids.
  • A final common pathway in the angiogenic response involves integrin-mediated information exchange between a proliferating vascular endothelial cell and the extracellular matrix. This class of adhesion receptors, called integrins, are expressed as heterodimers having an α and P subunit on all cells. One such integrin, αvβ3, is the most promiscuous member of this family and allows endothelial cells to interact with a wide variety of extracellular matrix components. Peptide and antibody antagonists of this integrin inhibit angiogenesis by selectively inducing apoptosis of the proliferating vascular endothelial cells. Two cytokine-dependent pathways of angiogenesis exist and may be defined by their dependency on distinct vascular cell integrins, αvβ3, and αvβ5. Specifically, basic FGF- and VEGF-induced angiogenesis depend on integrin αvβ3 and αvβ5, respectively, since antibody antagonists of each integrin selectively block one of these angiogenic pathways in the rabbit corneal and chick chorioallantoic membrane (CAM) models. Peptide antagonists that block all α, integrins inhibit FGF- and VEGF-stimulated angiogenesis. While normal human ocular blood vessels do not display either integrin, αvβ3 and αvβ5 integrins are selectively displayed on blood vessels in tissues from patients with active neovascular eye disease. While only αvβ3 was consistently observed in tissue from patients with ARMD, αvβ3 and αvβ5 both were present in tissues from patients with proliferative diabetic retinopathy. Systemically administered peptide antagonists of integrins blocked new blood vessel formation in a mouse model of retinal vasculogenesis.
  • Hence, anti-angiogenic agents have a role in treating retinal degeneration to prevent the damaging affects of these trophic and growth factors. Angiogenic agents, also have role in promoting desirable vascularization to retard retinal degeneration by enhancing blood flow to cells.
  • Immense research efforts have contributed to our understanding of the mechanism of angiogenesis during disease progression, and as a result of these studies, a large number of angiostatic molecules have been, or are currently being, tested in clinical trials. However, to date, the results from these clinical trials have been disappointing, and the benefits from these antiangiogenic treatments in patients have been minimal at best.
  • Many factors may require consideration before angiostatic therapies ultimately become successful. Naturally occurring compensatory mechanisms may ultimately render angiogenic monotherapies obsolete. Angiostatic drugs generally target a single cytokine or intracellular angiogenic pathway. In vivo, angiogenesis is likely to be initiated by the combined signaling of multiple pathways. Thus, blocking a single pathway may be insufficient to prevent angiogenesis during the treatment of neovascular diseases. Further complicating matters, it is also likely that blocking a single pathway induces compensation and increased roles of other angiogenic pathways.
  • WO 03/080648 relates to recombinant adeno-associated viral (rAAV) vector compositions comprising nucleic acid segments encoding therapeutic gene products, and their use in the manufacture of medicaments for treating various disorders of the eye including, for example, retinal, ocular or choroidal neovascularization (CNV).
  • It has now been discovered that a concurrent administration of a combination of angiostatic compounds that target different pathways enhances angiostatic potency and also interferes with natural compensatory mechanism.
  • Summary of the Invention
  • The present invention relates to compositions which an useful in a method of treating a neovascular disease, such as a retinal neovascular disease, by administering to a mammal suffering from a neovascular disease an amount of a combination of angiogenesis suppressing drugs sufficient to inhibit new blood vessel formation.
  • In a first aspect, the present invention provides a composition comprising (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1 having the formula:
    Figure imgb0001
    The angiostatic fragment of TrpRS preferably has the amino acid residue sequence of SEQ ID NO: 1. The composition may further comprise at least one therapeutic agent selected from the group consisting of an angiostatic steroid, an anti-neoplastic agent, an anti-bacterial agent, an anti-viral agent, and an anti-inflammatory agent. Preferably, the angiostatic fragment ofTrpRS is a dimer.
  • In one embodiment, the composition of the first aspect of the present invention is for treating a neovascular disease.
  • In a second aspect, the present invention provides the use of (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1 having the formula:
    Figure imgb0002
    for the preparation of a pharmaceutical composition for treatment of a neovascular disease in a mammal by administering a vascular development inhibiting amount of a combination of drugs comprising (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1. The disease may be a retinal neovascular disease. Preferably, the combination of drugs is to be administered by intravitreal injection into the eye of a mammal suffering from a retinal neovascular disease, preferably selected from the group consisting of an ischemic retinopathy, a vascular hemorrhage, a vascular leakage, a choroidopathy, age related macular degeneration, diabetic retinopathy, presumed ocular histoplasmosis, retinopathy of prematurity, sickle cell anemia, and retinitis pigmentosa.
  • In a third aspect, the present invention provides the use of (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1 having the formula:
    Figure imgb0003
    for the preparation of a pharmaceutical composition for treatment of a tumor in a mammal by administering a vascular development inhibiting amount of a combination of drugs comprising (i) an angiostatic fragment of TrpRS wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting ofSEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) Compound 1. In one embodiment, the mammal is a human. Preferably, the angiostatic fragment of TrpRS has the amino acid residue sequence of SEQ ID NO: 1.
  • In a fourth aspect, the present invention provides a pharmaceutical composition comprising a composition of the first aspect of the present invention and a pharmaceutically acceptable carrier therefor.
  • Neovascular diseases treatable by the compositions of the present invention include, without limitation, ocular diseases such as retinal degenerative diseases, retinal vascular degenerative diseases, ischemic retinopathies, vascular hemorrhages, vascular leakage, and choroidopathies in neonatal, juvenile, or fully mature mammals. The compositions of the present invention can also be utilized to treat neovascular diseases such as solid tumor cancers (e.g., lung cancer, breast cancer, and prostate cancer) and rheumatoid arthritis, for example.
  • Brief Description of the Drawings
    • Figure 1 depicts the amino acid sequences of angiostatic fragments of tryptophanyl-tRNA synthetase designated as T2-TrpRS, SEQ ID NO: 1 and T2-TrpRS-GD, SEQ ID NO: 2 (a mutant thereof).
    • Figure 2 depicts the amino acid sequence of angiostatic fragments of tryptophanyl-tRNA synthetase designated as mini-TrpRS, SEQ ID NO: 3 and T1-TrpRS, SEQ ID NO: 4.
    • Figure 3 depicts the amino acid sequence of full length TrpRS (SEQ ID NO: 5) and indicates the position of the T1, T2 and Mini fragments thereof.
    • Figure 4 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 1 intravitreally injected with PBS.
    • Figure 5 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Examples 1 intravitreally injected with (A) 0.5x concentration (10 mg/ml) of peptidomimetic integrin signaling inhibitor Compound (1); (B) a 1x concentration (2 mg/ml) of VEGF aptamer Compound (2); and (C) a combination of integrin signaling inhibitor Compound (1) and VEGF aptamer Compound (2).
    • Figure 6 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 2 intravitreally injected with phosphate buffered saline (PBS).
    • Figure 7 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 2 intravitreally injected with 0.1x concentration (0.05 mg/ml) of T2-TrpRS.
    • Figure 8 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 2 intravitreally injected with 0.1x concentration of VEGF aptamer Compound (2).
    • Figure 9 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 2 intravitreally injected with a combination of 0.1x concentration of T2-TrpRS and 0.1x concentration of VEGF aptamer Compound (2).
    • Figure 10 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 3 intravitreally injected with PBS.
    • Figure 11 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 3 intravitreally injected with a 1x concentration of T2-TrpRS.
    • Figure 12 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 3 intravitreally injected with a 1x concentration of VEGF aptamer Compound (2).
    • Figure 13 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 3 intravitreally injected with a 1x concentration of T2-TrpRS and a 1x concentration of VEGF aptamer Compound (2).
    • Figure 14 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 4 intravitreally injected with phosphate buffered saline (PBS).
    • Figure 15 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 4 intravitreally injected with a 1x concentration of T2-TrpRS.
    • Figure 16 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 4 intravitreally injected with a 0.5x concentration of VEGF aptamer Compound (2).
    • Figure 17 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 4 intravitreally injected with a combination of a 1x concentration of T2-TrpRS and 0.5x concentration of VEGF aptamer Compound (2).
    • Figure 18 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 5 intravitreally injected with PBS.
    • Figure 19 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 5 intravitreally injected with a 1x concentration of T2-TrpRS.
    • Figure 20 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 5 intravitreally injected with a 0.5x concentration ofpeptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 21 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 5 intravitreally injected with a combination of a 1x concentration of T2-TrpRS and a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 22 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 6 intravitreally injected with PBS.
    • Figure 23 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 6 intravitreally injected with a 1x concentration of T2-TrpRS.
    • Figure 24 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 6 intravitreally injected with a 1x concentration of VEGF aptamer Compound (2).
    • Figure 25 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 6 intravitreally injected with a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 26 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 6 intravitreally injected with a combination of a 1x concentration of T2-TrpRS and a 1x concentration of VEGF aptamer Compound (2).
    • Figure 27 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 6 intravitreally injected with a combination of a 1x concentration of T2-TrpRS and a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 28 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 6 intravitreally injected with a combination of a 1x concentration of T2-TrpRS, a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1), and a normal concentration of VEGF aptamer Compound (2).
    • Figure 29 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 7 intravitreally injected with PBS.
    • Figure 30 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 7 intravitreally injected with a 1x concentration of T2-TrpRS.
    • Figure 31 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 7 intravitreally injected with a 1x concentration of VEGF aptamer Compound (2).
    • Figure 32 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 7 intravitreally injected with a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 33 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 7 intravitreally injected with a combination of a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1) and a 1x concentration of VEGF aptamer Compound (2).
    • Figure 34 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 7 intravitreally injected with a combination of a 1x concentration of T2-TrpRS, a 0.5x concentration of peptidomimetic integrin signaling inhibitor Compound (1), and a 1x concentration of VEGF aptamer Compound (2).
    • Figure 35 depicts photomicrographs of primary and secondary vascular layers of retinas of control mice from Example 9 intravitreally injected with PBS.
    • Figure 36 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a 1x concentration of peptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 37 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a 1x concentration of VEGF aptamer Compound (2).
    • Figure 38 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a 1x concentration of T2-TrpRS.
    • Figure 39 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a 1x concentration of each of T2-TrpRS and peptidomimetic integrin signaling inhibitor Compound (1).
    • Figure 40 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a 1x concentration of each of peptidomimetic integrin signaling inhibitor Compound (1) and VEGF aptamer Compound (2).
    • Figure 41 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a combination of a 1x concentration of each of T2-TrpRS and VEGF aptamer Compound (2).
    • Figure 42 depicts photomicrographs of primary and secondary vascular layers of retinas of mice from Example 9 intravitreally injected with a combination of a 1x concentration of each of the inhibitors T2-TrpRS, peptidomimetic integrin signaling inhibitor Compound (1), and VEGF aptamer Compound (2).
    • Figure 43 is a graphical representation of data from a T2-TrpRS dosing experiment.
    • Figure 44 is a graphical representation of data from a VEGF-aptamer dosing experiment.
    • Figure 45 is a graphical representation of data showing inhibition of deep vascular plexus formation as a function of administered optimal concentration of VEGF aptamer and T2-TrpRS compound, alone and in combination.
    • Figure 46 is a graphical representation of data from αvβ3 and αvβ5 integrin antagonist dosing experiment.
    • Figure 47 is a graphical representation of data showing inhibition of deep vascular plexus formation as a function of administered optimal concentration of a small molecule αvβ03 and αvβ5 integrin antagonist and T2-TrpRS, alone and in combination.
    • Figure 48 is a graphical summary of data showing inhibition of deep vascular plexus formation at various combinations of therapies.
    • Figure 49 is a graphical representation of data showing degree of inhibition of deep vascular plexus formation at various combinations of therapies.
    • Figure 50 is a series of photomicrographs of primary and deep (secondary) vascular layers at various therapies and combinations thereof at dosing levels shown in Figure 49.
    • Figure 51 is a graphical representation of levels of inhibition of vascular plexus formation with triple combination therapy at various dosage levels.
    • Figure 52 is a graphical representation similar to Figure 51 but showing inhibition of >75%, >90% and 100%.
    • Figure 53 is a graphical representation of levels of inhibition of vascular plexus formation comparing monotherapies versus combination therapies at various dosage levels.
    • Figure 54 is a graphical representation of >75%, >90% and 100% levels of inhibition of vascular plexus formation comparing monotherapies versus combination therapies at various dosage levels.
    • Figure 55 is a graphical representation of data showing an area of neovascular tufts as a function of various monotherapies as well as a triple combination therapy utilizing a single injection of a therapeutic agent or agents.
    • Figure 56 is similar to Figure 55, but shows data from a dual injection of a therapeutic agent or agents.
    • Figure 57 is a graphical representation of data showing areas of neovascular tufts as a function of various monotherapies, dual therapies and a triple therapy.
    • Figure 58 is a series of photomicrographs showing mouse retinas treated with angiostatic compounds singly and in combination.
    • Figure 59 shows the structure of VEGF aptamer Compound (2) (SEQ ID NO: 6), pegaptanib sodium.
    • Figure 60 shows a graph of survival for tumor bearing rats treated with a composition of the invention (squares) versus control rats treated only with PBS (triangles).
    Detailed Description of Preferred Embodiments
  • The angiostatic fragments of TrpRS are a 43 kDa fragment (i.e., the T2 fragment, "T2-TrpRS", SEQ ID NO: 1, or a mutant of T2-TrpRS, "T2-TrpRS-GD", SBQ ID NO: 2; both of which are shown in Figure 1), the 48 kDa fragment known as mini-TrpRS (SBQ ID NO: 3, shown in Figure 2), and the 46 kDa fragment known as T1-TrpRS (SEQ ID NO: 4, shown in Figure 2). The amino acid residue sequence of T2-TrpRS-GD (SEQ ID NO: 2), differs from SEQ ID NO: 1 by two amino acid residue substitutions (i.e., S121G and Y122D). The amino acid residue sequence of full length human TrpRS (SEQ ID NO: 5) is shown in Figure 3, along with an indication of the position of the T1, T2 and mini fragments thereof. Without being bound by theory, it is believed that the angiostatic fragments of TrpRS can form noncovalent dimers (see e.g., Yu et al. J. Biol. Chem. 2004, 279: 8378-8388), which may contribute to the biological activity of the fragments. Accordingly, any reference herein an in the appended claims to an angiostatic fragment of TrpRS (e.g., T1-TRpRS, T2-TrpRS, mini-TrpRS) is to be construed as a reference to the monomer form, the dimer form, or a mixture thereof.
  • The compound having the formula of Compound (1) is available from Merck KGaA (Darmstadt, Germany) as EMD 472523 and is a peptidomimetic integrin signaling inhibitor.
  • Compound (1)
  • Figure imgb0004
    The VEGF signaling inhibitor pegaptanib sodium (Compound (2)) is a nuclease resistant, 2'-fluoropyrimidine RNA-based aptamer specific for VEGF-165 and is a polyethoxylated oligonucleotide having the following formula (SEQ ID NO: 6; Figure 59, R in Figure 59 is a 40 kiloDalton polyethylene glycol (PEG) chain): 5'-40K PEG-C5 aminolinker-CfGmGmArArUfCfAmGmUfGmAmAmUfGmCfUf UfAmUfAmCfAmUfCfCfCm3' - 3'dT
    wherein
  • Cf = 2'fluoro C
    Ar = 2' OH (ribo) A
    Uf = 2'fluoro U
    3' - 3'dT = inverted deoxyT
    Am = 2'OMe A
    C5 aminolinker = pentyl amino linker
    Gm = 2'OMe G
    40K PEG = 40K polyethelene glycol amide.
  • A polyethoxylated oligonucleotide of SEQ ID NO: 6 is sold commercially under the trademark MACUGEN® by from Eyetech Pharmaceuticals, Inc., and is also known as NX1838 or pegaptanib sodium.
  • In one embodiment the combination of drugs also includes at least one additional therapeutic agent such as an angiostatic steroid, an anti-neoplastic agent, an anti-bacterial agent, an anti-viral agent, an anti-inflammatory agent, and the like.
  • Examples of suitable angiostatic steroids include anecortave acetate and triamcinolone acetonide.
  • Examples of suitable anti-neoplastic agents include Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthranaycin; Asparaginase; Asperlin ; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer, Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefingol; Chlorambucil; Cirolemycia ; Cisplatin; Cladribine; Crisnatol Mesylata; Cyclophosphamide; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin Hydrochloride; Decitabine; Dexormaplatin; Dezaguanine; Dezaguanine Mesylate; Diaziquone; Docetaxel; Doxorubicin; Doxombicin Hydrochloride; Droloxifene; Droloxifene Citrate; Dramostanolone Propionate; Duazomycin, Edatrexate; Eflornithine Hydrochloride; Elsamitrucin; Enloplatin; Enpromate; Epipropidine; Epirubicin Hydrochloride; Erbulozole; Esorubicin Hydrochloride; Estramustine; Estramustine Phosphate Sodium; Etanidazole; Ethiodized Oil I 131; Etoposide; Etoposide Phosphate; Etoprine; Fadrozole Hydrochloride; Fazarabine; Fenretinide; Floxuridine; Fludarabine Phosphate; Fluorouracil; Flurocitabine; Fosquidone; Fostriecin Sodium; Gemcitabine; Gemcitabine Hydrochloride; Gold Au 198; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide; Imofosine; Interferon Alfa-2a; Interferon Alfa-2b ; Interferon Alfa-n1; Interferon Alfa-n3; Interferon BetarIa; Interferon Gamma-Ib; Iproplatin; Irinotecan Hydrochloride; Lanreotide Acetate; Letrozole; Leuprolide Acetate Liarozole Hydrochloride; Lometrexol Sodium; Lomustine; Losoxantrone Hydrochloride; Masoprocol; Maytansine; Mechlorethamine Hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Metoprine; Meturedepa; Mitindomide; Mitocarcin; Mitocromin; Mitogillin; Mitomalcin; Mitomycin; Mitosper; Mitotane; Mitoxantrone Hydrochloride; Mycophenolic Acid; Nocodazole; Nogalamycin; Ormaplatin; Oxisuran; Paclitaxel; Pegaspargase; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Plicamycin; Plomestane; Porfimer Sodium; Porfiromycin; Prednimustine; Procarbazine Hydrochloride; Puromycin; Puromycin Hydrochloride; Pyrazofurin; Riboprine; Rogletimide; Safingol; Safingol Hydrochloride; Semustine; Simtrazene; Sparfosate Sodium; Sparsomycinl; Spirogermanium Hydrochloride; Spiromustine; Spiroplatin; Streptonigrin; Streptozocin; Strontium Chloride Sr 89; Sulofenur, Talisomycin; Taxane; Taxoid; Tecogalan Sodium; Tegafur, Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testolactone; Thiamiprine; Thioguanine; Thiotepa; Tiazofurin; Tirapazamine; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Taciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Verteporfin; Vinblastina Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate; Vinepidine Sulfate; Vinglycinaba Sulfate; Vinleurosine Sulfate; Vinorelbine Tartrate; Vinrosidine Sulfate; Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; and Zornbicin Hydrochloride.
  • Example of suitable anti-bacterial agents include, but are not limited to, penicillins, aminoglycosides, macrolides, monobactams, rifamycins, tetracyclines, chloramphenicol, clindamycin, lincomycin, imipenem, fusidic acid, novobiocin, fosfomycin, fusidate sodium, neomycin, polymyxin, capreomycin, colistimethate, colistin, gramicidin, minocycline, doxycycline, vanomycin, bacitracin, kanamycin, gentamycin, erythromicin and cephalosporins.
  • Examples of suitable anti-inflammatory agents include, but are not limited to, aspirin (acetylsalicylic acid), indomethacin, sodium indomethacin trihydrate, salicylamide, naproxen, colchicine, fenoprofen, sulindac, diflunisal, diclofenac, indoprofen and sodium salicylamide.
  • Examples of suitable anti-viral agents include, but are not limited to, alpha-methyl-P-adamantane methylamine, 1-D-ribofuranosyl-1,2,4-triazole-3 carboxamide, 9-(2 hydroxy-ethoxy)methylguavine, adamaatanamine, 5-iodo-2'-deoxyuiidine, trifluorothymidine, interferon, adenine arabinoside, CD4, 3'-azido-3'-deoxythymidine (AZT), 9-(2-hydroxyethoxymethyl)-guanine (acyclovir), phosphonofomic acid, 1-adamaatanamine, peptide T, and 2',3'-dideoxycytidine.
  • Neovascular diseases treatable by the compositions of the present invention include, without limitation, neovascular diseases of the eye (e.g., retinal and choroidal neovascular diseases), rubeotic glaucoma, pterygia, solid tumor cancers (e.g., lung cancer, breast cancer, and prostate cancer), osteoarthritis, rheumatoid arthritis, vascular anomalies and malformations (e.g., hemangiomas, lymphangiomas, and the line), and psoriasis.
  • The compositions of the present invention may be used in a method of treating retinal neovascular diseases in a mammal. Preferably, the method comprises intravitreally injecting into the eye of a mammal suffering from a neovascular disease a vascular development inhibiting amount of a combination of antiangiogenic and angiostatic compositions that provide an angiostatic fragment of TrpRS as described above, the VEGF signaling inhibitor pegaptanib sodium; and the integrin signaling inhibitor compound (1).
  • This method can be utilized to treat ocular diseases such as vascular degenerative diseases, ischemic retinopathies, vascular hemorrhages, vascular leakage, and choroidopathies in neonatal, juvenile or fully mature mammals. Examples of such diseases include age related macular degeneration, diabetic retinopathy, presumed ocular histoplasmosis, retinopathy of prematurity, sickle cell anemia, hemangioma, pterygia, ischemic central retinal vein occlusion, blanch retinal vein occlusion, ocular melanoma, retinal blastoma, and retinitis pigmentosa, as well as retinal injuries.
  • Another aspect of the present invention is a therapeutic composition useful for the treatment of neovascular diseases, which comprises an angiostatic fragment of TrpRS as described above, the VEGF signaling inhibitor pegaptanib sodium, and the integrin signaling inhibitor compound (1), together with one or more pharmaceutically acceptable excipient.
  • In a preferred embodiment, the composition further comprises at least one additional therapeutic agent such as an angiostatic steroid, an anti-neoplastic agent, an anti-bactenal agent, an anti-viral agent, an anti-inflammatory agent, and the like.
  • The compositions of the present invention may be used in a method of treating a neovascular disease comprising administering to a mammal suffering from a neovascular disease a vascular development inhibiting amount of a combination of drugs comprising an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) as described above, the vascular endothelial growth factor (VEGF) signaling inhibitor pegaptanib sodium and the integrin signaling inhibitor compound (1).
  • Generally, a vascular development inhibiting amount of a composition of the present invention is at least about 10 µg/kg body weight and, in most cases, not in excess of about 8 mg/kg body weight per day for systemic treatments. Preferably the dosage is in the range of about 10 µg/kg body weight to about 1 mg/kg body weight daily. For ocular, intravitreal treatment of human patients, the preferred dosage is in the range of about 0.1 to about 5 milligrams per eye for a given treatment. The compositions may be administered in a single dose or in multiple doses over time. One of ordinary skill in the medical arts would be capable of determining the optimum effective therapeutic dosage of a composition of the present invention, taking into account the particular patient, the drugs present in the composition, the disease state, and other factors that are well known in the medical arts.
  • The therapeutic compositions of this invention can be embodied in a variety of physical forms. These forms include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspensions, aerosols, liposomes, suppositories, injectable and infusible solutions and sustained release forms. One of ordinary skill in the medical arts will choose a suitable dosage form depending on the intended mode of administration and on the disease to be treated, using pharmacological principles well known in the art.
  • A therapeutic composition according to this invention can be administered by conventional routes of administration, such as parenteral, subcutaneous, intravenous, intramuscular, intralesional, intrasternal, intravitreal, intracranial, or aerosol routes. Topical routes of administration can also be used, with application of the compositions locally to a particular part of the body (e.g., eye, skin, lower intestinal tract, vagina, rectum) where appropriate. The therapeutic compositions also include conventional pharmaceutically acceptable carriers and excipients that are known to those of skill in the art.
  • Generally, the therapeutic compositions of the present invention can be formulated and administered using methods and compositions similar to those used for the individual classes of active ingredients present in the compositions. It will be understood by those of skill in the art that conventional doses will vary depending upon the particular active ingredients in the composition, as well as the patient's health, weight, age, sex, the condition or disease, and the desired mode of administration.
  • The therapeutic compositions of this invention include pharmacologically appropriate, pharmaceutically acceptable carriers, excipients and vehicles. In general, these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media such as phosphate buffered saline (PBS). Parenteral vehicles can include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. In addition, intravenous vehicles can include fluid and nutrient replenishers, and electrolyte replenishers, such as those based on Ringer's dextrose. Excipients such as preservatives and other additives can also be present, such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases. Suitable formulation aids, carriers, other excipients, and methods of formulating pharmaceutical compositions are disclosed in Remington's Pharmaceutical Sciences, 14th Ed., Mack Publishing Co., 1970, particularly Part VIII, "pharmaceutical Preparations and Their Manufacture", pages 1461 - 1762.
  • The therapeutic compositions of the present invention can be packaged in suitably sterilized bottles or vials, either in multi-dose or in unit dose forms. The containers are preferably hermetically sealed after being filled with a composition of the invention. Preferably, the compositions are packaged in a container having a label affixed thereto, which label identifies the drugs present in the composition, and bears a notice in a form prescribed by a government agency such as the United States Food and Drug Administration, reflecting approval of the composition under appropriate laws, dosage information, and the like. The label preferably contains information about the composition that is useful to a health care professional administering the composition to a patient. The package also preferably contains printed informational materials relating to the administration of the composition, instructions, indications, and any necessary required warnings.
  • METHODS Neonatal Mouse Retinal Angiogenesis Model
  • Description of the Model. Immediately after birth (postnatal day zero; "P0"), retinal vasculature is virtually absent in the mouse. By four weeks after birth (P28) the retina has attained an adult pattern of retinal vessels coincident with the onset of vision. Physiological neovascularization of the retina occurs during this period via a stereotypical, biphasic developmental pattern of angiogenesis. During the primary phase of retinal vascular development, spoke-like peripapillary vessels grow radially from the central retinal artery and vein, becoming progressively interconnected by a capillary plexus that forms between them. This "inner retinal plexus" grows in area, volume and complexity, centrifugally, as a monolayer within the nerve fiber layer during the first seven to ten days following birth.
  • The second phase of retinal vessel formation begins between postnatal days 7 (P7) and 10 (P10) when collateral branches sprout from capillaries of the superficial plexus and penetrate into the retina where their tips branch and anastamose laterally to form a planar "deep vascular plexus". While the deep vascular plexus is in place by P14, it undergoes extensive remodeling from P14 to P21. It is of interest to note that the formation of these vascular networks in the neonatal mouse are strikingly similar to the events occurring in the third trimester human fetus.
  • Advantages and Quantification of the Model. The reproducibility of the murine retinal development process and its easy accessibility in neonatal animals provide an opportunity to assess the efficacy of antiangiogenic compounds in a physiologically relevant model of angiogenesis. Additional advantages of the neonatal mouse model are the ability to qualitatively and quantitatively evaluate the angiostatic effect of putative antagonists of angiogenesis. Angiostatic activity was evaluated based upon the degree of angiogenesis in the deep, outer retinal vascular layer (secondary layer) that develops between P8 and P12. The appearance of the inner blood vessel network (primary layer) was evaluated for normal development and signs of toxicity. No abnormalities in the inner vascular layer were observed in any of the assays performed and described herein. Qualitative evaluation of the secondary layer vascularization can be performed by microscopically photographing appropriately stained superficial and deep layers of excised retinas and determining the percentage of eyes in which formation of the deep vascular layer is completely or partially inhibited. All data presented herein are based on qualitative analysis of the percentage of eyes that demonstrated a 75 to 100 % inhibition of deep retinal vascular network formation after treatment. In most cases, the percentage of mice that exhibited > 95 % and 100 % inhibition of deep retinal vascular network formation are also provided.
  • Preparation of compositions. Peptidomimetic integrin signaling inhibitor Compound (1) is solubilized in PBS at a concentration of about 20 mg/ml in PBS (1x concentration). T2-TrpRS is solubilized in PBS at a concentration of about 0.5 mg/ml (1x concentration). VEGF aptamer (pegaptanib sodium; Compound (2)) is solubilized in PBS at a concentration of about 2 mg/ml (1x concentration) to achieve an injection of approximately 1 µg/eye at 1x concentration. For all combination assays the compounds were prepared at 2 or 3 times the 1x concentration of each material and then combined to produce a final solution containing each individual compound at the same concentration as was used alone (e.g., 1x, 0.5x, 0.25x or 0.1x, as the case may be). For all assays, a single injection of 0.5 µl of PBS solutions of the drugs was administered intravitreally regardless of the number of compounds being injected. As used herein, the term "0.1x" refers to one tenth of the 1x concentration of a given material, "0.25x" refers to one quarter of the 1x concentration of a given material, "0.5x" refers to one half of the 1x concentration of a given material, and so forth for similar designations.
  • Mouse model of Oxygen Induced Retinopathy (OIR). This model is described by Smith, L., Invest. Ophthalmol. Vis. Sci. 35, 101-111 (1994). Mice are placed in hyperoxia (75% O2) from P7-P12, followed by return to normoxia. While under hyperoxia, the central retinal vessels obliterate, and deep vasculature fails to form. Upon return to normoxia, the retina becomes hypoxic and pathological neovascularization results.
  • Quantification of neovascularization in the OIR model involved quantification of neovascular tuft formation, as well as quantification of obliteration. Retinal whole mounts are prepared and blood vessels thereof are stained with isolectin GS-IB4. Confocal imaging, focusing just above the superficial vascular plexus, is carried out and a montage of four quadrants is made. Neovascular tufts are identified (Adobe PHOTOSHOP®) and the area of pixelation is quantified In addition, areas of obliteration are traced (Adobe PHOTOSHOP®), and the area of pixelation is quantified. Conversion factor based on image acquisition (resolution, size etc.) is then applied to obtain a value in µm2.
  • General Angiogenesis Assay Procedure.
  • An in vivo angiogenesis assay in the neonatal mouse (Balb/C, The Jackson Laboratory, Bar Harbor, ME) was used to evaluate the angiostatic activity of integrin signaling inhibitor Compound (1), T2-TrpRS, and VEGF aptamer Compound (2). Intravitreous injection and retina isolation was performed with a dissecting microscope (SMZ 645, Nikon, Japan). An eyelid fissure was created at postnatal day 7 (P7) with a fine blade to expose the globe for injection. The samples (0.5 µl) were injected with a Hamilton syringe fitted with a 32-gauge needle (Hamilton Company, Reno, NV). The injection was made between the equator and the corneal limbus. During injection, the location of the needle tip was monitored by direct visualization to determine that it was in the vitreal cavity. Eyes with needle-induced lens or retinal damage were excluded from the study. After the injection, the eyelids were repositioned to close the fissure.
  • On postnatal day 12 (P12), animals were euthanized and the eyes were enucleated. After about 10 minutes in 4% paraformaldehyde (PFA) the cornea, lens, sclera, and vitreous were excised through a limbal incision. The isolated retina was prepared for staining by soaking in methanol for about 10 minutes on ice, followed by blocking in 50% fetal bovine serum (Gibco, Grand Island, NY) with 20% normal goat serum (The Jackson Laboratory, Bar Harbor, ME) in PBS for about one hour on ice. The blood vessels were specifically visualized by staining the retina for about 18 hours at about 4 °C with a rabbit antimouse collagen IV antibody (Chemicon, Temecula, CA) diluted 1:200 in blocking buffer or with a fluorescent conjugated isolectin (Griffonia simplicifolia, Molecular Probes). An ALEXA FLUOR® (Alexa) 594-conjngated goat anti-rabbit IgG antibody (Molecular Probes, Eugene, OR) (1:200 dilution in blocking buffer) was incubated with the retina for about 2 hours at about 4°C. The retinas were then mounted for microscopic evaluation with slow-fade mounting media (Molecular Probes, Eugene, OR).
  • Example 1. Treatment of neonatal mouse eyes with a combination of a peptidomimetic integrin signaling inhibitor and a VEGF signaling inhibitor.
  • Following the General Angiogenesis Assay Procedure ("General Procedure") described hereinabove, the eyes of neonatal Balb/C mice were intravitreally injected on postnatal day 8 (P8) with either a 0.25x concentration of integrin signaling inhibitor Compound (1) (five mice), a 0.5x concentration of VEGF aptamer Compound (2) (five mice), or a combination of a 0.25x concentration Compound (1) and a 0.5x concentration of Compound (2) (six mice). As a control, another group of six mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 1 and in Figures 4 and 5. Table 1.
    % inhibition: 0-10% 10-25% 25-50% 50-75% 75-100%
    PBS
    100 0 0 0 0
    0.5x Compound (2) 20 20 20 20 40
    0.1x Compound (1) 40 0.5 20 20 20
    Combination 0 17 0 0 83
  • Example 2. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 0.1x concentration of T2-TrpRS (eight mice), a 0.1x concentration of VEGF aptamer Compound (2) (eight mice), or a combination of a 0.1x concentration T2-TrpRS and a 0.1x concentration of Compound (2) (ten mice). As a control, another group of eight mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 2 and in Figures 6, 7, 8, and 9. Table 2.
    % inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS 100.0 0 0 0 0 0 0
    0.1x Comp. (2) 87.5 12.5 0 0 0 0 0
    0.1x T2-TrpRS 50 37.5 12.5 0 0 0 0
    Combination 10 30 20 20 20 0 0
  • Example 3. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS (eight mice), a 1x concentration of VEGF aptamer Compound (2) (eight mice), or a combination of a 1x concentration T2-TrpRS and a 1x concentration of Compound (2) (ten mice). As a control, another group of six mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 3 and in Figures 10, 11, 12, and 13. Table 3.
    % inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS
    100 0 0 0 0 0 0
    1x Comp. (2) 37.5 12.5 12.5 12.5 25 0 0
    1x T2-TrpRS 12.5 12.5 0 12.5 62.5 12.5 12.5
    Combination 0 0 10 10 80 40 20
  • Example 4. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS (eight mice), a 0.5x concentration of VEGF aptamer Compound (2) (ten mice) or a combination of a 1x concentration T2-TrpRS and a 0.5x concentration of Compound (2) (ten mice). As a control, another group of six mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 4 and in Figures 14, 15, 16, and 17. Table 4.
    % inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS 66.6 16.7 16.7 0 0 0 0
    0.5x Comp. (2) 60 30 0 0 10 0 0
    1x T2-TrpRS 12.5 37.5 12.5 12.5 25 0 0
    Combination 0 0 10 10 70 30 20
  • Example 5. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and an integrin signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 0.5x concentration of integrin signaling inhibitor Compound (1) or a combination of a 1x concentration T2-TrpRS and a 0.5x concentration of Compound (1), in groups of six mice for each treatment regimen. As a control, another group of four mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 5 and in Figures 18, 19, 20, and 21. Table 5.
    % inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS
    50 25 25 0 0 0 0
    0.5x Comp. (1) 50 33.3 16.7 0 0 0 0
    1x T2-TrpRS 33.3 16.7 33.3 0 16.7 16.7 0
    Combination 0 33.3 16.7 0 50 16.7 0
  • Example 6. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 0.5x concentration of integrin signaling inhibitor Compound (1), a 1x concentration of VEGF aptamer Compound (2), a combination of a 1x concentration T2-TrpRS and a 0.5x concentration of Compound (1), a combination of a 1x concentration T2-TrpRS and a 1x concentration of Compound (2), or a combination of a 1x concentration T2-TrpRS, a 0.5x concentration of Compound (1), and a 1x concentration of Compound (2), in groups of eight mice for each treatment regimen. As a control, another group of eight mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 6 and in Figures 22, 23, 24, 25, 26, 27, and 28. Table 6.
    % Inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS
    100 0 0 0 0 0 0
    0.5x Comp. (1) 50 12.5 12.5 12.5 12.5 0 0
    1 x Comp. (2) 37.5 25 12.5 25 0 0 0
    1x T2-TrpRS 50 25 12.5 12.5 0 0 0
    T2-TrpRS + Comp. (2) 25 25 25 0 25 25 12.5
    T2-TrpRS + Comp. (1) 50 0 0 0 50 37.5 25
    T2-TrpRS + (1) and (2) 0 0 0 0 100 87.5 75
  • Example 7. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 0.5x concentration of integrin signaling inhibitor Compound (1), a 1x concentration of VEGF aptamer Compound (2), a combination of a 0.5x concentration Compound (1) and a 1x concentration of Compound (2), or a combination of a 1x concentration T2-TrpRS, a 0.5x concentration of Compound (1), and a 1x concentration of Compound (2), in groups of eight mice for each treatment regimen. As a control, another group of six mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 7 and in Figures 29, 30, 31, 32, 33, and 34. Table 7.
    % Inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS
    100 0 0 0 0 0 0
    0.5x Comp. (1) 12.5 25 12.5 12.5 37.5 0 0
    1x Comp. (2) 25 25 0 12.5 37.5 0 0
    1x T2-TrpRS 25 12.5 12.5 0 50 12.5 0
    Comp. (1) + (2) 0 12.5 12.5 25 50 25 25
    T2-TrpRS + (1) and (2) 0 0 0 0 100 62.5 50
  • Example 8. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS and a VEGF signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS (ten mice), a 0.25x concentration of VEGF aptamer Compound (2) (eleven mice), or a combination of a 1x concentration T2-TrpRS and a 0.25x concentration of Compound (2) (eleven mice). As a control, another group of eight mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 8. Table 8.
    % inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS
    100 0 0 0 0 0 0
    0.25x Comp.(2) 54.5 36.4 0 0 9.1 0 0
    1x T2-TrpRS 30 30 20 10 10 0 0
    Combination 18.2 18.2 9.1 18.2 27.3 0 0
  • Example 9. Treatment of neonatal mouse eyes with a combination of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor.
  • Following the General Procedure, the eyes of neonatal Balb/C mice (in groups of eight mice each) were intravitreally injected on P4 with either a 1x concentration of T2-TrpRS, a 1x concentration of integrin signaling inhibitor Compound (1), a 1x concentration of VEGF aptamer Compound (2), a combination of a 1x concentration T2-TrpRS and a 1x concentration of Compound (1), a combination of a 1x concentration Compound (1) and a 1x concentration of Compound (2), or a combination of a 1x concentration T2-TrpRS, a 1x concentration of Compound (2), and a 1x concentration of Compound (2). As a control, another group of eight mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 9 and Figures 35-42. Table 9.
    % Inhibition: 0-10% 10-25% 25-50% 50-75% 75-100% >95% 100%
    PBS
    75 25 0 0 0 0 0
    1x Comp. (1) 25 12.5 12.5 37.5 12.5 0 0
    1x Comp. (2) 25 0 12.5 12.5 50 25 12.5
    1x T2-TrpRS 12.5 0 25 12.5 50 12.5 0
    Comp. (1)+T2-TrpRS 0 12.5 12.5 25 50 25 12.5
    Comp. (1) + (2) 12.5 12.5 0 12.5 62.5 62.5 50
    T2-TrpRS + Comp. (2) 0 12.5 12.5 12.5 62.5 37.5 25
    T2-TrpRS + (1) + (2) 0 25 0 0 75 75 62.5
  • Example 10. Treatment of neonatal mouse eyes with varying doses of an angiostatic fragment of TrpRS.
  • Following the General Procedure, the eyes of neonatal Balb/C mice (in groups of eight to twelve mice each) were intravitreally injected on P4 with T2-TrpRS, at concentrations of 0.1x (8 mice), 0.3x (12 mice), 1x (12 mice), 2x (12 mice), and 3x (12 mice). As a control, another group of 10 mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 10. Table 10.
    % Inhibition: 0-10% 10-25% 25-50% 50-75% 75-100%
    PBS
    100 0 0 0 0
    0.1 T2-TrpRS 75 25 0 0 0
    0.3 x T2-TrpRS 33.3 16.7 8.3 16.7 25
    1 x T2-TrpRS 8.3 16.7 25 8.7 41.7
    2x T2-TrpRS 8.3 25 0 16.7 41.7
    3x T2-TrpRS 66.7 25 8.7 0 0
  • Example 11. Treatment of neonatal mouse eyes with varying doses of an angiostatic fragment of TrpRS.
  • Following the General Procedure, the eyes of neonatal Balb/C mice (in groups of six to fourteen mice each) were intravitreally injected on P4 with T2-TrpRS, at concentrations of 0.3x (6 mice), 1x (14 mice), 2x (8 mice), 3x (7 mice), and 5x (6 mice). As a control, another group of 10 mice received only an intravitreal injection of PBS. At P12, the mice were euthanized and the retinas were removed from the injected eyes, stained, mounted and microscopically evaluated as described in the General Procedure. The vascularity of the secondary (outer retinal vascular) layer was evaluated based on the percentage of vascularization compared to the control eyes. The results are shown in Table 11. Table 11.
    % Inhibition: 0.10% 10-25% 25-50% 50-75% 75-100%
    PBS
    60 40 0 0 0
    0.3 x T2-TrpRS 33.3 16.7 0 0 50
    1x T2-TrpRS 14.3 14.3 7.1 14.3 50
    2xT2-TrpRS 12.5 12.5 12.5 12.5 50
    3x T2-TrpRS 14.3 14.3 143 28.5 28.5
    5x T2-TrpRS 16.6 333 333 16.6 0
  • The data for mice exhibiting >95 % and 100 % inhibition in the foregoing Examples demonstrates that even compositions comprising at least two materials selected from the group consisting of an angiostatic fragment of TrpRS, a VEGF signaling inhibitor, and an integrin signaling inhibitor afford unexpectedly greater efficacy for inhibition of neovascularization in the neonatal mouse eye model than the expected levels of inhibition from the simple additive effects of the combination of individual components. This is also evident when the results of the various examples are combined as in Table 12, which compiles the results from the Examples at concentrations of 1x to 2x of integrin inhibitor Compound (1), 0.5x to 1x of VEGF aptamer Compound (2), and 1x of T2-TrpRS, as well as compositions (inhibition values in bold type in Table 12) comprising combinations of at least two of Compound (1), Compound (2) and T2-TrpRS. The number of mice in each group is indicated in parenthesis for each grouping. The data in Table 12 clearly shows an unexpectedly higher level of inhibition of blood vessel formation in the deep vascular layer for mouse eyes treated with the compositions comprising combinations of at least two of Compound (1), Compound (2) and T2-TrpRS compared to the inhibition levels of the treatments with the individual inhibitors by themselves or the numerical sum thereof. Table 12. Composite Data from the Examples.
    %Inhibition: 0-10% 16-15% 15-50% 50-75% 75-100% >95% 100%
    PBS (38 mice) 84.2 10.5 53 0 0 0 0
    1x-2x Comp. (1) (30 mice) 33.3 20 133 16.7 16.7 0 0
    1x Comp. (2) (30 mice) 39.1 20 7.6 11.9 21.4 4.7 2.4
    1x 77-TrpRS (46 mice) 23.9 17.4 15.2 8.7 34.8 8.7 2.2
    Comp. (1)+T2-TrpRS (22 mice) 18.2 13.6 9.1 9.1 50 27.3 13.6
    Comp. (1) + (2) (1 6 mice) 6.3 12.5 6.3 18.8 56.3 43.7 37.5
    T2-TrpRS + Comp. (2) (36 mice) 5.6 11.1 139 8.3 61.1 33.3 19.4
    T2-TtpRS+(1)+(2) (24 mice) 0 8.3 0 0 91.7 79.2 62.5
  • The results from Examples 10 and 11 (Tables 10 and 11) show that the efficacy of T2- TrpRS reached a maximum at about 1x to 2x concentration and did not provide any more than 50% of mice having 75-100% inhibition in the deep vascular layer. The efficacy began to fall off at 2x and greater concentration. Thus even with increasing dosages, T2 did not provide greater than about 50% efficacy at the 95-100 % Inhibition leveL
  • Example 12. Synergistic effects of the administration of T2-TrpRS angiostatic fragment, VEGF aptamer and peptidomimetic αvβ3 and αvβ5 integrin signaling inhibitor.
  • To study the effect of combining different angiostatic molecules, three angiostatic compounds known to target critical, but separate, angiogenic pathways were utilized: a small molecule integrin αvβ3 and αvβ5 antagonist (Compound (1); "EMD 472523" obtained from Merck KGaA, Darmstadt, Germany), a VEGF165 antagonist (Compound (2); pegaptanib sodium), and a truncated form of Tryptophan tRNA synthetase (T2-TrpRS, obtained from Angiosyn Inc., La Jolla, CA). Although the exact mechanism of action for T2-TrpRS has not been fully elucidated, its mechanism of action is not directly linked to VEGF or integrin antagonism.
  • The neonatal mouse retinal angiogenesis model was used to test the efficacy of each monotherapy, and various combinations of these individual compounds. As stated hereinabove, mice are born without a retinal vasculature. During the first three weeks after birth an adult-like vasculature develops. The retinal vasculature forms three distinct planar plexuses with the superficial vascular plexus developing during the first post-natal week. At post-natal day 8 (P8), the vessels of the superficial plexus branch and migrate toward the deep plexus at the outer edge of the inner nuclear layer. To test the angiostatic properties of each monotherapy or combination solution, intravitreal injections were performed at P7, when the formation of the superficial network is nearing completion, but before the formation of the deep plexuses has begun. The effects on formation of the deep vascular plexus were then analyzed five days later (P12). The degree of inhibition for each injected retina was scored as 0-10%, 10-25%, 25-50%, 50-75%, or 75-100% (Figure 48) with the 75-100% inhibition group further separated into >90% and 100% inhibition levels (Figure 49). The appearance of the previously formed primary vascular plexus, as well as the overall retinal morphology, were evaluated for signs of toxicity.
  • Sample preparation. The αvβ3 and αvβ5 integrin antagonist (Compound (1)) was stored as lyophilized powder in a desiccator at room temperature (I.A.) or -20°C (V.A.), and solubilized in sterile, RNAse-free 1x PBS immediately before use. The VEGF aptamer (Compound (1)) was synthesized as a 40 kDa PEG conjugated compound (Transgenomic Inc, Boulder, CO) based on published information, Bridonneau, et al., J. Chromatogr: B. Diomed. Sci. App. 726: 237-47 (1999). The compound was determined to be pure by reverse phase liquid chromatography. Concentrations reported herein refer to the final concentration of the active VEGF aptamer rather than the total concentration of the PEG conjugated compound and were determined by spectrophotometric analysis at 260/280 nm. T2-TrpRS peptide was made as a recombinant compound as described in Otani, et al., Proc. Nat'l. Acad. Sci., USA 99: 178-183 (2002) and U.S. Provisional Application for Patent Serial No. 60/598,019 filed August 2, 2004 . Purified product was stored in 50% glycerol at -20 °C, and dialyzed into sterile 1x PBS immediately before use. Combination solutions were prepared by initially creating 3x stock solutions of each individual compound. The compounds were then mixed together, and with PBS where appropriate, to make a final solution containing each desired compound at a concentration equivalent to each corresponding monotherapy concentration.
  • Intravitreal injections. All animal work adhered to strict protocol guidelines for the humane care and use of animals. Intravitreal injections were performed, the retinas were dissected, and the vasculature was visualized. OIR was induced according to the protocol described by Smith, et al., Invest. Ophthalmol. Vis. Sci., 35: 101-111 (1994), by exposing post-natal day 7 (P7) pups and their mothers to an environment of 75% oxygen (hyperoxia) for 5 days, followed by a return to room air (normoxia). Intravitreal injections were performed at P12, immediately following return to normoxia and the retinas were analyzed at P 17. Blood vessels were stained using isolectin GS-IB4 from Griffonia simplicifolia (lectin GS), conjugated to Alexa Fluor 594 (Molecular Probes, 1:150 dilution in PBS). Confocal images were taken using a 4x objective lens, carefully focusing just above the inner limiting membrane of the retina, making the pre-laminar neovascular tufts prominent. Four overlapping images were acquired from each retina and each individual image was converted to a 2 x 2 inch size with 300 pixels per inch. The neovascular tuft areas were quantified by masked individuals using retinal whole mounts. Tufts were specifically selected, based on their characteristic appearance and the higher intensity of isolectin staining, using the magic wand tool in Adobe PHOTOSHOP® software. The total area in pixels was then determined. The areas of neovascular tuft formation were normalized to PBS-injected control OIR retinas.
  • Dosing. Dosing experiments were first performed to determine the maximum efficacy dose for each individual compound. Each compound was found to have a bell-shaped efficacy curve with optimal effective doses of 5-10 µg (10-20 nmoles) per eye for the integrin antagonist (Figure 46), 1.0-2.0 µg (108-215 pmoles) per eye for the VEGF aptamer (Figure 44), and 0.25-0.5 µg (5.2-10.4 pmoles) per eye for T2-TrpRS (Figure 43). Single injections of each monotherapy at the optimal dose, and solutions containing appropriate combinations of each compound at equivalent doses were then performed to compare the aagiostatic activities. At the maximum individual doses, around 35% of the retinas were unaffected by injection of either the integrin antagonist or the VEGF aptamer. The other 67% of the retinas basically fell evenly within the 10-25%, 25-50%, 50-75%, or 75%-100% range (Table 13A below). Inhibition of the deep vascular network with T2-TrpRS, peptide was slightly better. 24% of the T2-TrpRS peptide injected retinas developed a normal, complete deep vascular plexus while 35% of the T2-TrpRS peptide injected retinas exhibited over 75% inhibition compared to 17% and 21% for the integrin antagonist and the VEGF aptamer respectively (Table 13A below). When the angiostatic compounds were injected in combination, the angiostatic effects on neovascularization were striking. Each dual combination, integrin antagonist + T2-TrpRS, integrin antagonist + VEGF aptamer, and T2-TrpRS + VEGF aptamer, demonstrated significant improvement of angiostatic activity over the monotherapies. Significantly fewer retinas were resistant to angiostatic treatment using the combination therapies. Neovascularization was inhibited by over 75% in a majority of the retinas treated with any of the dual combinations (Table 13 A below).
  • When all three compounds were injected together at the same optimal doses as the corresponding monotherapy injections (1x triple combination), over 90% of the retinas had > 75% inhibition. Unlike the monotherapy or double combination-treated retinas, all retinas treated with the triple combination exhibited some degree of neovascular inhibition. In addition, only 8% of the injected retinas still had any significant levels of neovascularization at all. Nearly complete inhibition of angiogenesis was observed in the other 92% of the retinas injected with the triple combination compound (Table 13A below; Figure 48). The differences in angiostatic efficacy became even more pronounced when the >75% inhibition category was subcategorized into >90% inhibition and 100% inhibition levels (Figure 49). Over 80% of the retinas injected with the triple combination had greater than 90% inhibition of deep vascular plexus formation and 63% had 100% inhibition of neovascularization where not even a single neovascular sprout could be observed. This is a substantial improvement over both the monotherapies which demonstrated 100% inhibition in < 5% of the treated retinas and the dual combination therapies. In addition, the superficial vascular plexus of many of the triple combination-treated retinas resembled that of a normal P7 retina rather than P12 retinas, indicating that further vascular growth within the superficial plexus had also been prevented by the triple combination immediately following injection. Inhibition of superficial plexus growth was not observed in any mono- or dual-therapy treated retina. The more central vessels of the superficial vascular plexus that had already formed prior to injection remained normal, indicating negligible levels of toxicity to pre-existent vasculature. In addition, no signs of neuronal toxicity were observed, and the retinal morphology was unaltered, indicating that no observable negative side effects had occurred by injection of the triple combination solution. Images of the superficial and deep vascular plexuses from one complete representative experiment are shown in Figure 50.
  • To analyze synergism, and to test if potent angiostatic activity could be maintained using lower doses of the triple combination, serial dilutions were tested. The triple combination was still highly effective at inhibiting angiogenesis when diluted up to 100-fold (0.01x triple combination) (Table 13B below; Figures 52 and 53). When the triple combination was made by combining the individual compounds at one-tenth their optimal dose, nearly 80% of the treated retinas still exhibited >75% inhibition, and 50% of the retinas exhibited complete (100%) inhibition of neovascularization. At the 0.1x concentrations (1 µg/eye integrin antagonist, 0.2 µg/eye VEGF aptamer, and 0.025 µg/eye T2-TrpRS), inhibition of neovascularization by the individual angiostatic compounds was negligible (Table 13C below; Figure 54). Some efficacy was observed after injection of the double 0.1x T2-TrpRS and 0.1x VEGF aptamer combination. However, despite the fact that this combination was the most effective angiostatic of all the double combinations tested, the angiostatic activity was still minimal compared to the inhibition levels observed by injection of the 0.1x triple combination. TABLE 13
    A. Neonatal mouse angiogenesis model combination experiment
    Percentage of retinas with the indicated levels of neovascular inhibition
    Injection N 0-10% 10-25% 25-50% 50-75% >75% >90% 100%
    PBS
    38 84.2 10.5 5.3 0.0 0.0 0.0 0.0
    5-10 µg integrin antagonist 30 33.3 20.0 13.3 16.7 16.7 0.0 0.0
    1-2 µg VEGF aptamer 42 39.1 20.0 7.6 11.9 21.4 4.7 2.4
    0.25 µg T2-TtpRS 46 23.9 17.4 15.2 8.7 34.8 8.7 2.2
    77-TrpRS + integrin ant. 22 18.2 13.6 9.1 9.1 50.0 27.3 13.6
    Integrin ant. + VEGF apt. 21 6.3 12.5 6.3 18.8 56.3 43.7 28.5
    T2-TrpRS + VEFG apt. 36 5.6 11.1 13.9 8.3 61.1 38.3 19.4
    Triple combination 24 0 8.3 0 0 91.7 83.2 62.6
    B. Triple combination serial dilution experiment
    Injection N 0-10% 10-25% 25-50% 50-75% >75% >96% 100%
    PBS
    14 71.4 14.3 14.3 0.0 0.0 0.0 0.0
    1x Triple Combination 16 0.0 0.0 0.0 7.1 92.8 71.4 57.1
    0.5x Triple Combination 16 0.0 0.0 0.0 0.5 100.0 87.5 50.0
    0.25x Triple Combination 16 0.0 0.0 0.0 0.5 100.0 68.8 43.8
    0.1x Triple Combination. 18 0.0 5.5 11.1 5.5 77.8 61.1 44.4
    0.05x Triple Combination 10 0.0 10.0 20.0 20.0 50.0 30.0 20.0
    0.01x Triple Combination 10 10.0 10.0 30.0 30.0 20.0 20.0 10.0
    C. Low dosing monotherapy vs. combination experiment
    Inhibition Levels: N 0-10% 10.25% 25-50% 50-75% >75% >90% 100%
    PBS
    8 100 0 0 0 0 0 0
    0.1x integrin ant. (1.0 µg) 10 90 10 0 0 0 0 0
    0.1x VEGF apt. (0.20 µg) 8 75 25 0 0 0 0 0
    0.1x T2-TrpRS (0.025 µg) 10 50 37.5 12.5 0 0 0 0
    0.1x T2-TrpRS+VEFG apt. 10 10 30 20.0 20 20 0 0
    0.1x Triple Combination 18 0 5.5 11.1 5.5 77.8 61.1 44.4
  • Example 13. Synergistic effects of a "Triple Therapy"
  • The mouse model of oxygen-induced retinopathy (OIR) described hereinabove is a well-accepted model of hypoxia-induced neovascularization in the retina. The associated vascular changes are consistent, reproducible and quantifiable. In recent years the use of this model has been extended to the general study of disease-related ischemic vasculopathies and related anti-angiogenic interventions. To study the synergistic properties of these angiostatic compounds in a more pathological model of angiogenesis, the effects of monotherapies and combination therapies on the formation of pathological neovascularization were tested in the mouse OIR model. For initial experiments, the optimal doses obtained from the neonatal angiogenesis model were used. In each case, combination therapies demonstrated improved angiostatic activities compared to the monotherapies. However, due to the angiostatic activities of each monotherapy, it was difficult to determine if the results of combining the various compounds were synergistic or simply additive. Thus, based on the results observed using the neonatal mouse retinal angiogenesis model which demonstrated equivalent efficacies of the combination therapies at relatively low doses, each monotherapy and the various combination therapies were tested at one-tenth of the optimal doses. Again, the concentration of each compound in the combination solutions was equivalent to the corresponding monotherapy concentration. At the lower concentrations, no significant inhibition of pathological neovascular tuft formation was observed following monotherapy treatments. However, significant reductions in neovascular tuft formations were observed using each double combination (Figure 57). When the integrin antagonist was combined with T2-TrpRS peptide, pathological tuft formation was reduced by > 50%. Combining the integrin antagonist with the VEGF aptamer reduced tuft formation by > 40%. When T2-TrpRS peptide was combined with the VEGF aptamer, Pathological neovascularization was reduced by nearly 80% compared to control-treated retinas. Many of the retinas treated with the double T2-TrpRS / VEGF aptamer combination looked nearly normal with virtually no pathological neovascularization evident (Figure 58).
  • A dramatic increase in angiostatic activity by combining multiple angiostatic compounds that target distinct Angiogenesis pathways has been demonstrated. Strong angiostatic activities were observed in both a developmental and a pathological model of angiogenesis even after combining the compounds at doses which have no monotherapeutic activity. This suggests a synergistic effect rather than simply an additive effect. This data also suggests that targeting multiple pathways may be required for effective clinical anti-angiogenic therapy and may provide a new paradigm for the treatment of neovascular diseases. Preferably, at least two anti-angiogenic therapies are combined (e.g., a combination of VEGF signaling inhibitor, such as a VEGF aptamer, combined with an angiostatic fragment of TrpRS, such as the T2 fragment of TrpRS, and optionally and integrin antagonist).
  • By targeting and inhibiting three separate angiogenic pathways, nearly complete inhibition of angiogenesis was obtained in two separate models of angiogenesis. The complete inhibition of neovascularization may be important for the effective treatment of angiogenesis-related diseases using angiostatic therapies. In our models, even the best results from monotherapy injections generally only blocked 50-75% of new vessel growth. This means that in most cases, a significant amount of neovascularization still developed. In contract, two-thirds of the neonatal mouse retinas injected with the triple combination therapy had complete 100% inhibition of neovascular formation (Table 13). Similarly, in the OIR model of pathological angiogenesis, a large portion of the treated mice demonstrated little or no pathological neovascular tuft formation (Figure 58). During cancer treatment, high levels of angiogenesis inhibition may be required to cause the complete starvation of tumor cells and prevent further tumor growth. Monotherapies that only inhibit 50% of neovascular growth are only likely to reduce, rather than eliminate, the oxygen and nutrients available to the rapidly growing tumor cells. Although this may initially slow growth, it may not be sufficient to prevent further tumor growth. In these instances, combination therapies that can achieve complete inhibition of neovascularization would greatly improve the results of anti-angiogenic therapies during cancer treatments. In addition, by using relatively low doses while maintaining strong angiostatic potential, the possibility of adverse side effects generated by angiostatic treatments can be minimized. Together, the foregoing data demonstrate the beneficial utility of combining different angiostatic molecules for the treatment of neovascularization associated with disease.
  • The compositions of the present invention, comprising an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS), a vascular endothelial growth factor (VBGF) signaling inhibitor, and an integrin signaling inhibitor, provide a new and surprisingly efficacious treatment regimen for neovascular diseases, particularly for neovascular diseases of the eye.
  • Example 14. Treatment of a tumor
  • Glioblastoma multiform is an incurable malignant brain tumor, usually fatal within one year of diagnosis. The 9L rat gliosarcoma cell line is used as a model for malignant gliomas. In both forms of glioma, the tumor is highly vascularized and infiltrates into normal brain tissue. Untreated animal receiving a bolus of 9L cells intracerebrally have a survival tie of approximately 3 weeks, once the tumor cells have been implanted. At day 0, intracerebral 9L tumors were established by stereotactic inoculation of about 50,000 cells in about 2 µl of Dulbecco's Modified Eagles's Medium (DMEM; Life Technologies, Gaithersburg, MD) into the right frontal lobe of CD 344 Fisher rats that had previously been anesthetized with ketamine and xylazine.
  • At day 6, a 10 µl bolus of a composition of the invention (4.5 mg/l of 17-TrpRS, 30 mg/l of Compound (1), and 6 mg/l of Compound (2); pegaptanib sodium) was injected stereotactically over the course of about 2 minutes into the same region of the brain as the 9L cells were implanted. Following injection of the bolus, a pump was inserted into a subcutaneous pocket between the scapulae. A catheter connected via tubing to the pump was inserted into the same burr hole made for introduction of the 9L cells, and was fixed in place. Each pump had a flow rate of about 8 µl per hour. An additional quantity of the composition of the invention was continuously pumped into the brain of each animal for about 24 hours. The continuous pumping distributed the composition throughout the entire hemisphere of the brain in which the tumor cells had been implanted. Nine rats received the composition of the invention and nine rats received straight PBS as a control group.
  • At day 13, an incision was made between the scapulae, and the pump was removed and replaced with an fresh pump. Treatment with the composition of the invention or PBS was resumed for an additional 24 hours with the new pump at the same pumping flow rates. There was a 21 percent increase in survival for rats treated with the composition of the invention compared to the PBS treated control group (See Figure 60).
  • SEQUENCE LISTING
    • <110> The Scripps Research Institute
    • <120> COMPOSITIONS AND METHODS FOR TREATMENT OF NEOVASCULAR DISEASES
    • <130> 18-209
    • <140> EP 05 804 888.5
      <141> 2005-06-06
    • <150> US 60/577,156
      <151> 2004-06-04
    • <150> US 60/585,273
      <151> 2004-07-01
    • <150> US 60/655,801
      <151> 2005-02-24
    • <160> 6
    • <170> FastSEQ for windows version 4.0
    • <210> 1
      <211> 379
      <212> PRT
      <213> homo sapiens
    • <400> 1
      Figure imgb0005
      Figure imgb0006
    • <210> 2
      <211> 379
      <212> PRT
      <213> homo sapiens
    • <400> 2
      Figure imgb0007
    • <210> 3
      <211> 424
      <212> PRT
      <213> homo sapiens
    • <400> 3
      Figure imgb0008
    • <210> 4
      <211> 401
      <212> PRT
      <213> homo sapiens
    • <400> 4
      Figure imgb0009
    • <210> 5
      <211> 471
      <212> PRT
      <213> homo sapiens
    • <400> 5
      Figure imgb0010
      Figure imgb0011
    • <210> 6
      <211> 27
      <212> DNA
      <213> Artificial sequence
    • <220>
      <223> Synthetic oligonucleotide
    • <220>
      <221> misc_feature
      <222> (1)..(1)
      <223> 2'fluoro C linked at position 5' via a pentyl amino linker to a 40 kiloDalton polyethylene glycol chain
    • <220>
      <221> misc_feature
      <222> (2), C3), (9), (11), (15)
      <223> 2'OMe G
    • <220>
      <221> misc_feature
      <222> (4), (5)
      <223> 2'OH (ribo) A
    • <220>
      <221> misc_feature
      <222> (6), (10), (14), (17), (18), (20), (24)
      <223> 2'fluoro U
    • <220>
      <221> misc_feature
      <222> (7), (16), (22), (25), (26)
      <223> 2'fluoro C
    • <220>
      <221> misc_feature
      <222> (8), (12), (13), (19), (21), (23)
      <223> 2'OMe A
    • <220>
      <221> misc_feature
      <222> (27)..(27)
      <223> 2'OMe G linked at position 3' to the 3' terminus of deoxyT
    • <400> 6
      cggaaucagu gaaugcuuau acauccg    27

Claims (13)

  1. A composition comprising (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) a compound having the formula:
    Figure imgb0012
  2. The composition of claim 1 wherein the angiostatic fragment of TrpRS has the amino acid residue sequence of SEQ ID NO: 1.
  3. The composition of claim 1 or 2 further comprising at least one therapeutic agent selected from the group consisting of an angiostatic steroid, an anti-neoplastic agent, an anti-bacterial agent, an anti-viral agent, and an anti-inflammatory agent.
  4. The composition of any one of claims 1 to 3 wherein the angiostatic fragment of TrpRS is a dimer.
  5. The composition of any one of claims 1 to 4 for use in a method of treating a neovascular disease.
  6. Use of (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) a compound having the formula:
    Figure imgb0013
    for the preparation of a pharmaceutical composition for treatment of a neovascular disease in a mammal by administering a vascular development inhibiting amount of a combination of drugs comprising (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) a compound having the formula:
    Figure imgb0014
  7. The use of claim 6 wherein the disease is a retinal neovascular disease.
  8. The use of claim 6 wherein the combination of drugs is to be administered by intravitreal injection into the eye of a mammal suffering from a retinal neovascular disease.
  9. The use of claim 8 wherein the disease is selected from the group consisting of an ischemic retinopathy, a vascular hemorrhage, a vascular leakage, a choroidopathy, age related macular degeneration, diabetic retinopathy, presumed ocular histoplasmosis, retinopathy of prematurity, sickle cell anemia, and retinitis pigmentosa.
  10. Use of (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) a compound having the formula:
    Figure imgb0015
    for the preparation of a pharmaceutical composition for treatment of a tumor in a mammal by administering a vascular development inhibiting amount of a combination of drugs comprising (i) an angiostatic fragment of tryptophanyl-tRNA synthetase (TrpRS) wherein the angiostatic fragment of TrpRS has an amino acid residue sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4; (ii) pegaptanib sodium; and (iii) a compound having the formula:
    Figure imgb0016
  11. The use of claim 6 or 10 wherein the mammal is a human.
  12. The use of any one of claims 6 to 11 wherein the angiostatic fragment of TrpRS has the amino acid residue sequence of SEQ ID NO: 1.
  13. A pharmaceutical composition comprising a composition of any one of claims 1 to 4 and a pharmaceutically acceptable carrier therefor.
EP05804888A 2004-06-04 2005-06-06 Compositions and methods for treatment of neovascular diseases Not-in-force EP1765362B1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
PL12002028T PL2484365T3 (en) 2004-06-04 2005-06-06 Compositions and method for treatment of neovascular diseases
EP12002027.6A EP2484364B1 (en) 2004-06-04 2005-06-06 Compositions for treatment of neovascular diseases
DK12002028.4T DK2484365T3 (en) 2004-06-04 2005-06-06 Compositions and Methods for the Treatment of Neovascular Diseases
PL05804888T PL1765362T3 (en) 2004-06-04 2005-06-06 Compositions and methods for treatment of neovascular diseases
EP12002028.4A EP2484365B1 (en) 2004-06-04 2005-06-06 Compositions and method for treatment of neovascular diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US57715604P 2004-06-04 2004-06-04
US58527304P 2004-07-01 2004-07-01
US65580105P 2005-02-24 2005-02-24
PCT/US2005/019800 WO2005117954A2 (en) 2004-06-04 2005-06-06 Compositions and methods for treatment of neovascular diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP12002027.6A Division EP2484364B1 (en) 2004-06-04 2005-06-06 Compositions for treatment of neovascular diseases

Publications (3)

Publication Number Publication Date
EP1765362A2 EP1765362A2 (en) 2007-03-28
EP1765362A4 EP1765362A4 (en) 2010-02-24
EP1765362B1 true EP1765362B1 (en) 2012-03-28

Family

ID=35463352

Family Applications (3)

Application Number Title Priority Date Filing Date
EP05804888A Not-in-force EP1765362B1 (en) 2004-06-04 2005-06-06 Compositions and methods for treatment of neovascular diseases
EP12002027.6A Not-in-force EP2484364B1 (en) 2004-06-04 2005-06-06 Compositions for treatment of neovascular diseases
EP12002028.4A Not-in-force EP2484365B1 (en) 2004-06-04 2005-06-06 Compositions and method for treatment of neovascular diseases

Family Applications After (2)

Application Number Title Priority Date Filing Date
EP12002027.6A Not-in-force EP2484364B1 (en) 2004-06-04 2005-06-06 Compositions for treatment of neovascular diseases
EP12002028.4A Not-in-force EP2484365B1 (en) 2004-06-04 2005-06-06 Compositions and method for treatment of neovascular diseases

Country Status (13)

Country Link
US (2) US7528106B2 (en)
EP (3) EP1765362B1 (en)
JP (1) JP4943324B2 (en)
KR (1) KR101224368B1 (en)
AT (1) ATE551060T1 (en)
AU (1) AU2005249586B2 (en)
CA (1) CA2568750C (en)
DK (2) DK1765362T3 (en)
ES (3) ES2450541T3 (en)
MX (1) MXPA06014092A (en)
PL (2) PL1765362T3 (en)
PT (2) PT1765362E (en)
WO (1) WO2005117954A2 (en)

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA06014092A (en) * 2004-06-04 2007-03-15 Scripps Research Inst Compositions and methods for treatment of neovascular diseases.
US20060024288A1 (en) * 2004-08-02 2006-02-02 Pfizer Inc. tRNA synthetase fragments
US8282921B2 (en) * 2004-08-02 2012-10-09 Paul Glidden tRNA synthetase fragments
WO2006091729A2 (en) * 2005-02-24 2006-08-31 The Scripps Research Institute Revascularization of ischemic retinal tissue and screening method therefor
ES2338400B1 (en) * 2008-05-06 2011-09-14 David Benet Ferrus SET OF ANTIANGIOGEN MOLECULES AND ITS USE.
CA2727622A1 (en) * 2008-06-11 2009-12-17 Atyr Pharma, Inc. Thrombopoietic activity of tyrosyl-trna synthetase polypeptides
WO2009158649A1 (en) 2008-06-26 2009-12-30 Atyr Pharma, Inc. Compositions and methods comprising glycyl-trna synthetases having non-canonical biological activities
US20120004185A1 (en) 2009-02-27 2012-01-05 Atyr Pharma, Inc. Polypeptide structural motifs associated with cell signaling activity
US8255830B2 (en) * 2009-03-16 2012-08-28 Apple Inc. Methods and graphical user interfaces for editing on a multifunction device with a touch screen display
DK2408905T3 (en) 2009-03-16 2017-08-28 Pangu Biopharma Ltd Compositions and Methods comprising Histidyl-tRNA Synthetase Splicing Variants with Non-Canonical Biological Activities
EP2414513B1 (en) 2009-03-31 2015-10-28 Atyr Pharma, Inc. Compositions and methods comprising aspartyl-trna synthetases having non-canonical biological activities
WO2011072266A2 (en) * 2009-12-11 2011-06-16 Atyr Pharma, Inc. Aminoacyl trna synthetases for modulating hematopoiesis
US8828395B2 (en) 2009-12-11 2014-09-09 Atyr Pharma, Inc. Antibodies that bind tyrosyl-tRNA synthetases
DK2509625T3 (en) 2009-12-11 2015-04-27 Atyr Pharma Inc Histidyl tRNA synthetases to reduce inflammation
WO2011139714A2 (en) 2010-04-26 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-trna synthetase
WO2011139801A2 (en) 2010-04-27 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl trna synthetases
US8961960B2 (en) 2010-04-27 2015-02-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
CN103097524B (en) 2010-04-28 2016-08-03 Atyr医药公司 The innovation for the treatment of, diagnosis and the antibody compositions relevant to the protein fragments of Alanyl-tRNA synthetase finds
AU2011248457B2 (en) 2010-04-29 2017-02-16 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl tRNA synthetases
US9068177B2 (en) 2010-04-29 2015-06-30 Atyr Pharma, Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
EP2563912B1 (en) 2010-04-29 2018-09-05 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl trna synthetases
AU2011248230B2 (en) 2010-05-03 2016-10-06 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-tRNA synthetases
JP6008840B2 (en) 2010-05-03 2016-10-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of phenylalanyl αtRNA synthetase
WO2011139988A2 (en) 2010-05-03 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of seryl-trna synthetases
CN103096925A (en) 2010-05-03 2013-05-08 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-tRNA synthetases
AU2011248101B2 (en) 2010-05-04 2016-10-20 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
EP2566492B1 (en) 2010-05-04 2016-10-26 aTyr Pharma, Inc. Innovative discovery of therapeutic compositions related to protein fragments of glutamyl-prolyl-trna synthetases
AU2011252990B2 (en) 2010-05-14 2017-04-20 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
CA2799480C (en) 2010-05-17 2020-12-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-trna synthetases
JP5906237B2 (en) 2010-06-01 2016-04-20 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of lysyl tRNA synthetase
NZ603813A (en) 2010-07-12 2015-03-27 Atyr Pharma Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-trna synthetases
US8999321B2 (en) 2010-07-12 2015-04-07 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
EP2593125B1 (en) 2010-07-12 2017-11-01 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-trna synthetases
CN103124561B (en) 2010-07-12 2017-05-03 Atyr 医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-trna synthetases
JP5964304B2 (en) 2010-08-25 2016-08-03 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of tyrosyl-tRNA synthetase
AU2011311956C1 (en) * 2010-10-06 2017-06-15 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases
US9714419B2 (en) 2011-08-09 2017-07-25 Atyr Pharma, Inc. PEGylated tyrosyl-tRNA synthetase polypeptides
US9816084B2 (en) 2011-12-06 2017-11-14 Atyr Pharma, Inc. Aspartyl-tRNA synthetases
WO2013086228A1 (en) 2011-12-06 2013-06-13 Atyr Pharma, Inc. Pegylated aspartyl-trna synthetase polypeptides
EP2797959A4 (en) 2011-12-29 2015-08-26 Atyr Pharma Inc Aspartyl-trna synthetase-fc conjugates
EP2814514B1 (en) 2012-02-16 2017-09-13 Atyr Pharma, Inc. Histidyl-trna synthetases for treating autoimmune and inflammatory diseases
ES2847383T3 (en) 2013-03-15 2021-08-03 Atyr Pharma Inc Fc-histidyl-tRNA synthetase conjugates
KR101899591B1 (en) * 2015-02-26 2018-09-17 재단법인 의약바이오컨버젼스연구단 Compositions for treating or preventing infection-induced inflammatory diseases and for enhancing immune responses comprising tryptophanyl-tRNA synthetase
EP3612215A4 (en) 2017-04-20 2021-05-26 aTyr Pharma, Inc. Compositions and methods for treating lung inflammation
WO2023001288A1 (en) * 2021-07-23 2023-01-26 百奥泰生物制药股份有限公司 Integrin gpiib/iiia antagonist and application thereof in combination with anti-vegf antibody

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4405378A1 (en) 1994-02-19 1995-08-24 Merck Patent Gmbh Adhesion receptor antagonists
US5753230A (en) 1994-03-18 1998-05-19 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
DE4415310A1 (en) 1994-04-30 1995-11-02 Merck Patent Gmbh Cyclopeptides
DE4429461A1 (en) 1994-08-19 1996-02-22 Merck Patent Gmbh Adhesion receptor antagonists
ATE170179T1 (en) 1994-11-02 1998-09-15 Merck Patent Gmbh ADHESION RECEPTOR ANTAGONISTS
DE19516483A1 (en) 1995-05-05 1996-11-07 Merck Patent Gmbh Adhesion receptor antagonists
US5767071A (en) 1995-06-07 1998-06-16 Ixsys Incorporated Sevenmer cyclic peptide inhibitors of diseases involving αv β3
US5780426A (en) 1995-06-07 1998-07-14 Ixsys, Incorporated Fivemer cyclic peptide inhibitors of diseases involving αv β3
NZ336035A (en) * 1996-11-05 2002-03-28 Childrens Medical Center Angiogenesis inhibitory composition comprising an inhibitory compound and antiinflammatory drug
US6559144B2 (en) 1997-02-13 2003-05-06 Merck Patent Gesellschaft Mit Bicyclic amino acids
DE19705450A1 (en) * 1997-02-13 1998-08-20 Merck Patent Gmbh Bicyclic aromatic amino acids
DE19755800A1 (en) 1997-12-16 1999-06-17 Merck Patent Gmbh Cyclopeptide derivatives
DE19831710A1 (en) 1998-07-15 2000-01-20 Merck Patent Gmbh New diacyl-hydrazine derivatives, are integrin inhibitors useful for treating e.g. thrombosis, cardiac infarction, tumors, osteoporosis, inflammation or infection
KR20010093303A (en) 1999-02-20 2001-10-27 플레믹 크리스티안 β-ALANINE DERIVATIVES
DE19916837A1 (en) 1999-04-14 2000-10-19 Merck Patent Gmbh New di- or tetrahydro-benzoazulene derivatives, are integrin inhibitors useful e.g. for treating thrombosis, coronary heart disease, arteriosclerosis, tumors, osteoporosis or rheumatoid arthritis
DE19932796A1 (en) 1999-07-14 2001-01-18 Merck Patent Gmbh Diacylhydrazine derivatives
DE19939980A1 (en) 1999-08-24 2001-03-01 Merck Patent Gmbh Inhibitors of the integrin alphavbeta¶3¶
WO2001075078A1 (en) * 2000-03-31 2001-10-11 The Scripps Research Institute HUMAN AMINOACYL-tRNA SYNTHETASE POLYPEPTIDES USEFUL FOR THE REGULATION OF ANGIOGENESIS
PT1381382E (en) * 2000-11-01 2009-03-03 Merck Patent Gmbh Methods and compositions for the treatment of diseases of the eye
US7229960B2 (en) * 2000-11-03 2007-06-12 University Of Vermont And State Agricultural College Methods and compositions for inhibiting GRB7
UA75898C2 (en) * 2000-11-14 2006-06-15 Merck Patent Gmbh Method for prophylaxis and treatment of eye diseases using antagonist of integrin receptors
IL152420A0 (en) * 2001-02-23 2003-05-29 Novartis Ag Novel oncolytic adenoviral vectors
NZ528193A (en) * 2001-02-23 2006-02-24 Scripps Research Inst Tryptophanyl-tRNA synthetase derived polypeptides useful for the regulation of angiogenesis
AU2002332430A1 (en) * 2001-07-26 2003-02-17 Novartis Ag Methods of treating neuropilin-mediated diseases
OA12720A (en) * 2001-11-09 2006-06-27 Eyetech Pharmaceuticals Methods for treating ocular neovascular diseases.
CA2479167A1 (en) * 2002-03-20 2003-10-02 University Of Florida Research Foundation, Inc. Raav vector compositions and methods for the treatment of choroidal neovascularization
CN100543036C (en) * 2002-05-06 2009-09-23 得克萨斯州大学系统董事会 The guiding protein matter of delivery treatments or diagnostic reagent
DK1660057T3 (en) * 2003-08-27 2012-08-20 Ophthotech Corp Combination therapy for the treatment of neovascular eye disorders
EP1711172A4 (en) * 2003-12-23 2008-07-16 Univ Pennsylvania Compositions and methods for combined therapy of disease
MXPA06014092A (en) * 2004-06-04 2007-03-15 Scripps Research Inst Compositions and methods for treatment of neovascular diseases.

Also Published As

Publication number Publication date
ES2382661T3 (en) 2012-06-12
JP2008501718A (en) 2008-01-24
US7528106B2 (en) 2009-05-05
JP4943324B2 (en) 2012-05-30
US20090285792A1 (en) 2009-11-19
EP2484364B1 (en) 2014-02-19
DK2484365T3 (en) 2013-12-02
EP1765362A2 (en) 2007-03-28
US20060003933A1 (en) 2006-01-05
PT2484365E (en) 2013-12-12
EP2484365A1 (en) 2012-08-08
PT1765362E (en) 2012-06-04
PL1765362T3 (en) 2012-08-31
KR20070041498A (en) 2007-04-18
EP1765362A4 (en) 2010-02-24
ES2450541T3 (en) 2014-03-25
EP2484365B1 (en) 2013-10-02
WO2005117954A3 (en) 2006-10-12
PL2484365T3 (en) 2014-03-31
WO2005117954A2 (en) 2005-12-15
CA2568750A1 (en) 2005-12-15
ATE551060T1 (en) 2012-04-15
KR101224368B1 (en) 2013-01-22
DK1765362T3 (en) 2012-05-14
MXPA06014092A (en) 2007-03-15
AU2005249586A1 (en) 2005-12-15
ES2439390T3 (en) 2014-01-22
EP2484364A1 (en) 2012-08-08
CA2568750C (en) 2014-05-27
AU2005249586B2 (en) 2010-12-23

Similar Documents

Publication Publication Date Title
EP1765362B1 (en) Compositions and methods for treatment of neovascular diseases
CN105209619B (en) MiR-204 and miR-211 and application thereof
Martin et al. Vitreous cefazolin levels after intravenous injection: effects of inflammation, repeated antibiotic doses, and surgery
US20060078553A1 (en) Diverse multi-unit complexes including a tRNA synthetase fragment
ES2382357T3 (en) Myeloid-type cells transfected for the treatment of retinopathy of prematurity and related diseases
EP4309659A1 (en) Complex for treating optic nerve disease, and preparation method therefor and use thereof
US20140079665A1 (en) Therapeutic anti-igf1r combinations
CN108601950A (en) The purposes of the risk of chemoprophylaxis with neuroprotection property or the ischemia reperfusion injury in reduction subject
EP2588122B1 (en) Perlecan domain v protects, repairs and restores ischemic brain stroke injury and motor function
Zayit-Soudry et al. Safety of intravitreal bevacizumab in the developing rabbit retina
ZA200700052B (en) Compositions and methods for treatment of neovascular diseases
CN103070877A (en) Vision care drug
Kerby et al. The effect of hydrocortisone and of piromen in vitro on leukocytes of patients receiving ACTH and cortisone therapy
EP2139510B1 (en) Treating cerebrovascular diseases with erythropoietin and granulocyte-colony stimulating factor jointly
US20060079672A1 (en) Kits for modulating angiogenesis
Szumny et al. Glaucoma-new possibilities, new research
EP1947115B1 (en) Combination of glycoisoforms for the treatment or prevention of septicemia, transgenic cell line that produces erythropoietin glycoisoforms, pharmaceutical composition comprising said combination, method of obtaining the cell line, method of producing the combination of glycoisoforms and methods for the treatment and prevention of septicaemia
TW202315886A (en) Novel mutant of recombinant ganoderma lucidum immunomodulatory protein and use thereof
CN116920125A (en) Application of CBS gene in preparation of diabetic retinopathy treatment drug
US20060078886A1 (en) Business methods for modulating angiogenesis
KR20240029777A (en) New mutants of recombinant reishi mushroom immunomodulatory protein and their uses
Pedro et al. Single Intravitreal Injection of Etamsylate for the Treatment of Geographic Atrophy Associated with Submacular Hemorrhage

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20061221

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR LV MK YU

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20100125

17Q First examination report despatched

Effective date: 20100512

RIC1 Information provided on ipc code assigned before grant

Ipc: A01N 43/04 20060101ALI20110803BHEP

Ipc: C07K 1/00 20060101ALI20110803BHEP

Ipc: A61K 31/70 20060101AFI20110803BHEP

Ipc: C07K 14/00 20060101ALI20110803BHEP

Ipc: C07K 17/00 20060101ALI20110803BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 551060

Country of ref document: AT

Kind code of ref document: T

Effective date: 20120415

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: SERVOPATENT GMBH

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602005033434

Country of ref document: DE

Effective date: 20120524

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20120528

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2382661

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20120612

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

LTIE Lt: invalidation of european patent or patent extension

Effective date: 20120328

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120629

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120728

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E014183

Country of ref document: HU

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120630

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20130103

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602005033434

Country of ref document: DE

Effective date: 20130103

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120628

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120328

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120606

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20150612

Year of fee payment: 11

Ref country code: CZ

Payment date: 20150512

Year of fee payment: 11

Ref country code: GB

Payment date: 20150603

Year of fee payment: 11

Ref country code: DE

Payment date: 20150602

Year of fee payment: 11

Ref country code: SE

Payment date: 20150611

Year of fee payment: 11

Ref country code: ES

Payment date: 20150512

Year of fee payment: 11

Ref country code: PT

Payment date: 20150603

Year of fee payment: 11

Ref country code: DK

Payment date: 20150610

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: HU

Payment date: 20150513

Year of fee payment: 11

Ref country code: PL

Payment date: 20150414

Year of fee payment: 11

Ref country code: IE

Payment date: 20150609

Year of fee payment: 11

Ref country code: AT

Payment date: 20150527

Year of fee payment: 11

Ref country code: BE

Payment date: 20150612

Year of fee payment: 11

Ref country code: NL

Payment date: 20150609

Year of fee payment: 11

Ref country code: FR

Payment date: 20150608

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20150625

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602005033434

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20160630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160606

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20160701

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 551060

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160606

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160607

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20161206

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20160606

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20170228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170103

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160607

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160606

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160701

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160606

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160606

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160606

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160606

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160607

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20181204